US11788147B2 - Markers of triple-negative breast cancer and uses thereof - Google Patents

Markers of triple-negative breast cancer and uses thereof Download PDF

Info

Publication number
US11788147B2
US11788147B2 US16/746,721 US202016746721A US11788147B2 US 11788147 B2 US11788147 B2 US 11788147B2 US 202016746721 A US202016746721 A US 202016746721A US 11788147 B2 US11788147 B2 US 11788147B2
Authority
US
United States
Prior art keywords
tnbc
pathway
subtype
genes
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US16/746,721
Other versions
US20200224282A1 (en
Inventor
Jennifer A. Pietenpol
Brian Lehmann
Josh Bauer
Xi Chen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Vanderbilt University
Original Assignee
Vanderbilt University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Vanderbilt University filed Critical Vanderbilt University
Priority to US16/746,721 priority Critical patent/US11788147B2/en
Assigned to VANDERBILT UNIVERSITY reassignment VANDERBILT UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEHMANN, Brian, BAUER, Josh, CHEN, XI, PIETENPOL, JENNIFER A
Publication of US20200224282A1 publication Critical patent/US20200224282A1/en
Application granted granted Critical
Priority to US18/381,126 priority patent/US20240158865A1/en
Publication of US11788147B2 publication Critical patent/US11788147B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • TNBC triple-negative breast cancer
  • ER estrogen receptor
  • PR progesterone receptor
  • HER2 human epidermal growth factor receptor 2
  • TNBC tumors are generally larger in size, are of higher grade, have lymph node involvement at diagnosis, and are biologically more aggressive (Haffty B G, et al. J Clin Oncol. 2006; 24(36):5652-5657).
  • presurgical (neoadjuvant) chemotherapy TNBC patients have a higher rate of distant recurrence and a poorer prognosis than women with other breast cancer subtypes (Haffty B G, et al. J Clin Oncol. 2006; 24(36):5652-5657; Dent R, et al. Clin Cancer Res. 2007; 13(15 pt 1):4429-4434).
  • TNBC Tumor cell lines lacking functional BRCA1 or BRCA2 are sensitive to PARP inhibitors in preclinical studies (Farmer H, et al. Nature. 2005; 434(7035):917-921). Clinical trials using both PARP inhibitors and DNA-damaging agents (e.g., cisplatin) in TNBC are currently underway and show promise in BRCA1/2-mutant tumors (Fong P C, et al. N Engl J Med. 2009; 361(2):123-134). Other studies identifying molecular markers of TNBC, such as VEGF (Burstein H J, et al. J Clin Oncol. 2008; 26(11):1810-1816), EGFR (Nielsen T O, et al. Clin Cancer Res.
  • TNBC triple negative breast cancer
  • methods of determining a triple negative breast cancer (TNBC) subtype in an individual in need thereof comprising determining expression of one or more genes (e.g., presence of one or more mRNAs and/or protein encoded by gene) in one or more TNBC cells of the individual; and comparing the expression of the one or more genes in the TNBC cells with the expression of the one or more genes in a control.
  • Increased expression of one or more genes comprising one or more cell cycle genes, cell division genes, proliferation genes, DNA damage response genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC BL1 subtype.
  • Increased expression of one or more growth factor signaling genes, glycolysis genes, gluconeogenesis genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC BL2 subtype.
  • Increased expression of one or more immune cell signaling genes, cytokine signaling genes, antigen processing and presentation genes, core immune signal transduction genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC IM subtype.
  • Increased expression of one or more cell motility genes, extracellular matrix (ECM) receptor interaction genes, cell differentiation genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC M subtype.
  • TNBC MSL subtype Increased expression of one or more hormone regulated genes in the TNBC cells compared to the control indicates that the TNBC is a TNBC LAR type.
  • the methods are directed to methods of determining a treatment protocol for the triple negative breast cancer (TNBC) patient based on the TNBC subtype.
  • TNBC triple negative breast cancer
  • the methods are directed to predicting whether an individual (e.g., patient) will benefit from a (one or more) treatment for a particular TNBC subtype.
  • the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) subtype comprising contacting a cell line that is a model for the TNBC subtype of interest with an agent to be assessed; and determining viability of the cell line in the presence of the agent, wherein if the viability of the cell line decreases in the presence of the agent, then the agent can be used to treat the TNBC subtype of interest.
  • the TNBC subtype of interest can be a TNBC BL-1 subtype, a TNBC BL-2 subtype, a TNBC IM subtype, a TNBC M subtype, a TNBC MSL subtype or a TNBC LAR subtype.
  • FIGS. 1 A- 1 C Filtering GE data sets to identify TNBCs.
  • FIG. 1 B Histograms show the distribution and frequency of tumors using relative ER, PR, and HER2 GE levels (log 2) and bimodal fit to identify TN tumor samples. Dashed line indicates the expression value at the center of the positive expression peak used to select controls for C.
  • FIGS. 2 A- 2 E Identification of TNBC subtypes.
  • 2 B Consensus clustering displaying the robustness of sample classification using multiple iterations ( ⁇ 1000) of k-means clustering.
  • 2 C The CDF depicting the cumulative distribution from consensus matrices at a given cluster number (k).
  • 2 D The optimal cluster number is 7 at the point in which the relative change in area ( ⁇ ) under the CDF plot does not change with increasing k.
  • 2E Principal component analysis graphically depicting fundamental differences in GE between the TNBC clusters. The cluster (subtypes) are named as shown.
  • FIG. 3 GE patterns within TNBC subtypes are reproducible. Heat maps showing the relative GE (log 2, ⁇ 3 to 3) of the top differentially expressed genes (P ⁇ 0.05) in each subtype in the training set (left) and the same differentially expressed genes used to predict the best-fit TNBC subtype of the validation set (right). Overlapping gene ontology (GO) terms for top canonical pathways in both the training and validation sets as determined by GSE-A are shown to the right of the heat maps.
  • GO gene ontology
  • FIGS. 4 A- 4 F Basal-like TNBC subtypes have differential sensitivity to DNA-damaging agents.
  • IC50 values for TNBC cell lines treated with PARP inhibitors ( 4 A) veliparib, ( 4 C) olaparib, or ( 4 E) cisplatin for 72 hours. Error bars reflect SEM for 3 independent experiments. Black horizontal lines above various bars in the plots indicate cell lines that failed to achieve an IC50 at the highest dose of veliparib (30 ⁇ M), olaparib (100 ⁇ M), or cisplatin (30 ⁇ M). Cell lines that carry BRCA1 or BRCA2 mutations (pink) are displayed below the graph.
  • FIGS. 5 A- 5 D Differential sensitivity of the LAR TNBC subtype to AR and Hsp90 inhibitors.
  • Black bar above bicalutamide indicates cell lines that failed to achieve an IC50.
  • Heat map displays relative AR expression (log 2) across TNBC cell lines.
  • FIGS. 6 A- 6 D Mesenchymal-like TNBC subtypes are sensitive to dasatinib and NVP-BEZ235.
  • Cell lines that have PIK3CA mutations (red) or are deficient in PTEN (blue, circle indicates mutated) are displayed below the NVP-BEZ235 graph.
  • FIG. 7 Xenograft tumors established from TNBC subtypes display differential sensitivity to cisplatin, bicalutamide, and NVP-BEZ235.
  • Nude mice bearing established tumors (25-50 mm3) from basal-like (HCC1806 and MDA-MB-468), LAR (SUM185PE and CAL-148), or mesenchymallike (CAL-51 and SUM159PT) were treated with cisplatin (red), bicalutamide (purple), NVP-BEZ235 (green), or vehicle (blue) for approximately 3 weeks.
  • Serial tumor volumes (mm3) were measured at the indicated days. Each data point represents the mean tumor volume of 16 tumors; error bars represent SEM.
  • FIG. 8 Random distribution of TNBC subtypes found within datasels. The percent of tumors in each cluster is displayed across 14 studies that comprise the training dataset.
  • FIGS. 11 A- 11 B TNBC subtypes identified by IHC.
  • 11 B Heatmap displays hierarchical clustering of ERBB2, PGR, ESR1 and AR expression in the tumors identified by IHC. Color bar identifies the cluster associated with each tumor.
  • FIG. 12 TNBC subtypes Identified by IHC display similar GE patterns to the six TNBC subtypes. IHC TNBC clusters are shown in silhouette plot with relative GE for genes unique to the six TNBC subtypes shown below.
  • FIG. 13 Differential GE across TNBC subtypes. Heatmaps show relative GE (log 2, ⁇ 2 to 2) associated with proliferation, DNA damage response, myoepithelial genes, immune signal transduction, TGF ⁇ signaling, growth factor receptors, EMT, WNT signaling, stem-like, claudin (CL), angiogenesis, AR and of AR target genes across TNBC subtypes (as in FIG. 3 ).
  • FIGS. 14 A- 14 B TNBC tumor subtypes differentially stain for the proliferation marker Ki-67.
  • 14 A Representative micrographs from 20 tumors showing IHC staining of the proliferation marker, Ki-67, in tumors from different TNBC subtypes.
  • pCR pathologic complete response
  • FIG. 16 1M subtype Is enriched in Immune cell signaling. Heatmaps showing the relative GE (log 2, ⁇ 2 to 2) for genes involved in immune cell surface antigens, cytokines, immune cell signal transduction, complement, chemokine, and antigen presentation across TNBC subtypes (as in FIG. 3 ).
  • FIGS. 18 A- 18 B TNBC tumor subtypes differentially stain for AR by IHC.
  • 18 A IHC staining for AR from 20 tumors with representative samples from each TNBC subtype shown.
  • 18 B Dot plot showing the quantification of nuclear AR staining based on intensity and the percent of nuclei staining positive for AA in 20 tumors; Note, in some cases one dot represents overlapping dots from multiple tumors.
  • FIGS. 19 A- 19 B An androgen-inducible gene signature segregates LAR tumors. Hierarchical clustering was performed on both the ( 19 A) training and ( 19 ) validation TNBC tumor sel using a 559 androgen-inducible gene signature (Hayes M J et al. Clin Cancer Res. 2008; 14(13):4038-4044).
  • FIGS. 20 A- 20 B TNBC subtypes differentially correlate with the intrinsic molecular subtypes.
  • 20 A Heatmaps show relative GE (log 2, ⁇ 2 to 2) of luminal and basal markers of breast cancer across all TNBC subtypes.
  • FIGS. 21 A- 21 D TNBC subtypes differ in relapse-free survival and distant metastasis-free survival.
  • Kaplan-Meier plot showing ( 21 A) to-year RFS or ( 21 C) DMFS in TNBC subtypes.
  • FIGS. 23 A- 23 B Chromosomal Aberrations in TNBC cell lines.
  • 23 B Box plots depicting the average number of chromosomal rearrangements (translocations, inversions, and deletions) in basal-like vs. mesenchymal-like subtypes.
  • Chromosome mode and rearrangements were obtained from Departments of Pathology and Oncology, Univ of Cambridge (www.path.cam.ac.uk/ ⁇ pawefish/cell%20line%20catalogues/breast-cell-lines.htm). *P ⁇ 0.01 by unpaired t-test.
  • FIGS. 25 A- 25 C LAR cell lines depend on AR expression for colony formation.
  • 25 A Top panel depicts relative AR mRNA levels obtained from GE microarrays performed on TNBC cell lines (log 2, centered on 0). Color bar identifies TNBC subtype classification for each cell line (immunomodulatory shown in orange and unclassified shown in red). Immunoblots showing relative expression AR protein in TNBC cell lines, Hsp90 serves as a loading control.
  • 25 B AR expression 72 h following transfection with siRNA pools of non-targeting (NT) or targeting AR in MFM-223, MDA-MB-453 and SUM185PE cells. GAPDH expression serves as a loading control.
  • 25 C Colony formation of MFM-223, MDA-MB-453 and SUM185PE cells 14d following siRNA transfection of AR or non-targeting control (NT).
  • FIGS. 26 A- 26 B Identification of PIK3CA mutations in AR-positive TNBC cell lines and tumors.
  • 26 A Detection of PIK3CA 1047 mutation using digital droplet PCR.
  • VIC wild-type
  • FAM mutant
  • FIG. 27 AR-expressing TNBC cells display active PI3K pathway.
  • Heatmap displays relative protein levels (RPPA) of AR, AKT (S473 and T308), GSK3 ⁇ (S9 and S21) and PTEN across a large panel of TNBC cell lines.
  • RPPA relative protein levels
  • AKT S473 and T308
  • GSK3 ⁇ S9 and S21
  • PTEN PTEN across a large panel of TNBC cell lines.
  • Hierarchical clustering was performed on cell lines and reveal clusters of cell lines with active PI3K through PTEN loss or PIK3CA mutations.
  • FIGS. 28 A- 28 C- 2 AR-positive TNBC cell lines display active PI3K pathway and are sensitive to PI3K inhibitors.
  • 28 A Immunoblot displays relative levels of AR, p-AKT, p-S6 in AR-dependent prostate cancer (LNCaP), human mammary epithelial cells (HMEC) and LAR TNBC, with GAPDH serving as a loading control.
  • 28 B IHC-stained sections display cellular levels of AR, and p-AKT in cell lines from the same tissue microarray (TMA).
  • Bar graphs display the 50% inhibitory concentration (IC 50 ) for TNBC cell lines treated with single-agent pan PI3K inhibitors (BKM-120 and GDC-0941) ( 28 C- 1 ) or PI3K/mTOR inhibitors (NVP-BEZ235 or GDC-0980) ( 28 C- 2 ) for 72 h. Black bars above graphs indicate cell lines in which the IC 50 was not reached at maximal dose. Bar below indicates normal HMEC cells and cell lines carrying PIK3CA mutations.
  • FIGS. 29 A- 29 B Genetic targeting of AR increases the efficacy of the PI3K inhibitor GDC-0941 and PI3K/mTOR inhibitor GDC-0980.
  • 29 A Immunoblot displays decreased AR expression at 72 h following infection with two shRNAs targeting AR (shAR1 and shAR2) compared to infection with nontargeting shRNA (shNT) in three AR-positive TNBC cell lines. Actin levels below serve as loading controls.
  • FIGS. 30 A- 30 B Pharmacological targeting of AR with bicalutamide increases the efficacy of GDC0941 and GDC0980 in AR-expressing TNBC.
  • 30 A Line graphs show relative viability of AR-expressing cell lines treated with increasing doses of GDC-0941 (top) or GDC-0980 (bottom) alone or in combination with 25 ⁇ M bicalutamide (CDX). Dashed black line depicts the theoretical line of additivity of both drugs determined from the effect of bicalutamide alone and either GDC-0941 or GDC-0980 alone.
  • FIGS. 31 A- 31 B Combined inhibition of AR and PI3K increases apoptotic cell death in AR+TNBC cell lines.
  • 31 A Bar graphs display relative caspase 3/7 activity (RLU) normalized to viable cell number 48 hrs after treatment with vehicle, positive control (3 ⁇ M adriamycin, ADR), 50 ⁇ M bicalutamide (CDX) and 3 ⁇ M GDC-0941 or 1 ⁇ M GDC-0980 alone or in combination with CDX.
  • RLU relative caspase 3/7 activity
  • CDX 50 ⁇ M bicalutamide
  • 3 ⁇ M GDC-0941 or 1 ⁇ M GDC-0980 alone or in combination with CDX.
  • 31 B Histograms show representative cell cycle analysis of MDA-MB-453 cells treated with similar conditions as above. Indicated percentages represent the fraction of hypodiploid DNA (sub-G1), indicative of late stage apoptotic DNA fragmentation.
  • FIGS. 32 A- 32 C Simultaneous targeting of AR and PI3K decreases viability of AR-expressing cell lines grown in a 3-D forced suspension assay and inhibits in vivo xenograft growth.
  • 31 A Representative images display 3-D cell aggregates of MDA-MB-453 cells treated with increasing doses of GDC-0941 or GDC-0980 in the absence or presence of 25 ⁇ M bicalutamide (CDX).
  • Dashed black line depicts the theoretical line of additivity determined from the effect of bicalutamide alone and either GDC-0941 or GDC-0980 alone.
  • Nude mice bearing established tumors from AR-positive TNBC cell lines (CAL-148 and MDA-MB-453) were treated with either bicalutamide (CDX, black hashed line), NVP-BEZ235, GDC-0980 or with the combination of bicalutamide and either NVP-BEZ235 (blue hashed line) or GDC-0980 (red hashed line).
  • Serial tumor volumes (mm 3 ) were measured at the indicated days. Each data point represents mean tumor volume of 16 tumors; error bars represent SEM.
  • FIGS. 33 A- 33 B Detection of PIK3CA mutations by Sanger and digital droplet PCR.
  • 33 A Electropherograms display representative results from sanger sequencing of the E20 amplicon in a PIK3CA wild-type (top) or a H1047R PIK3CA mutant sample.
  • 33 B Scatterplot shows the percent of cells with mutant PIK3CA detected by digital droplet compared to Sanger sequencing.
  • FIG. 34 Genetic targeting of AR increases the efficacy of the PI3K inhibitor BKM-120 and PI3K/mTOR inhibitor NVP-BEZ235.
  • Line graphs display relative viability of LAR cell lines infected with nontargeting (shNT) or shRNAs targeting AR (shAR1 and shAR2) following a 72 h treatment with the pan-PI3K inhibitor BKM-120 (top) or the dual PI3K/mTOR inhibitor NVP-BEZ235.
  • FIG. 35 Pharmacological targeting of AR with bicalutamide increases the efficacy of BKM120 and NVP-BEZ235 in AR-expressing TNBC.
  • Line graphs show relative viability of AR-expressing cell lines treated with increasing doses of BKM120 (top) or NVP-BEZ235 (bottom) alone or in combination with 25 ⁇ M bicalutamide (CDX).
  • Dashed black line depicts the theoretical line of additivity determined from the effect of bicalutamide alone and either BKM120 or NVP-BEZ235 alone.
  • FIGS. 36 A- 36 B Simultaneous targeting of AR and PI3K decreases viability of AR-expressing cell lines grown in a 3-D forced suspension assay.
  • 36 A Line graphs display relative viability of 3-D cell aggregates treated with BKM120 or NVP-BEZ235 alone or in combination with 25 uM bicalutamide (CDX). Dashed black line depicts the theoretical line of additivity determined from the effect of bicalutamide alone and either BKM120 or NVP-BEZ235 alone.
  • 36 B Immunoblots from three AR-expressing TNBC cell lines treated with either bicalutamide (CDX), BKM-120, BEZ235 alone or in combination with bicalutamide for 24 h or 48 h. Western blots were probed for AR, PARP, p-AKT, AKT, p-S6, S6 and GAPDH.
  • FIG. 37 Combined Targeting of PI3K and AR increase apoptosis detected by annexin V and propidium iodide (PI) staining. Scattergrams show the percentage of viable (lower left quadrant, annexin V and PI negative), early apoptotic (lower right quadrant, annexin V-positive and PI-negative), or late apoptotic/necrotic (upper right quadrant, annevin V and PI positive) cells following 48 hrs of treatment with either GDC-0941 or GDC-0980 alone or in combination with 50 ⁇ M bicalutamide (CDX).
  • viable lower left quadrant, annexin V and PI negative
  • early apoptotic lower right quadrant, annexin V-positive and PI-negative
  • late apoptotic/necrotic upper right quadrant, annevin V and PI positive
  • FIGS. 38 A- 38 C Genetic and pharmacological targeting of AR increases p-AKT.
  • FIG. 39 PDGFR inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines.
  • Bar graph displays the half-maximal inhibitory concentration (nM) of the PDGFR inhibitor sorafenib for a panel of TNBC cell lines.
  • FIG. 40 IGF1R inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines.
  • Bar graph displays the half-maximal inhibitory concentration (nM) of the IGF1R inhibitor OSI-906 for a panel of TNBC cell lines.
  • FIG. 41 IGF1R inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines.
  • Bar graph displays the half-maximal inhibitory concentration (nM) of the IGF1R inhibitor BMS-754807 for a panel of TNBC cell lines.
  • FIG. 42 MET inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines.
  • Bar graph displays the half-maximal inhibitory concentration (nM) of the MET inhibitor PF2341066 for a panel of TNBC cell lines.
  • TNBC triple-negative breast cancer
  • GE gene expression
  • IM immunomodulatory
  • M mesenchymal
  • MSL mesenchymal stem-like
  • LAR luminal androgen receptor
  • Predicted “driver” signaling pathways were pharmacologically targeted in these cell line models as proof of concept that analysis of distinct GE signatures can inform therapy selection.
  • BL1 and BL2 subtypes had higher expression of cell cycle and DNA damage response genes, and representative cell lines preferentially responded to cisplatin.
  • M and MSL subtypes were enriched in GE for epithelial-mesenchymal transition, and growth factor pathways and cell models responded to NVP-BEZ235 (a PI3K/mTOR inhibitor) and dasatinib (an abl/src inhibitor).
  • the LAR subtype includes patients with decreased relapse-free survival and was characterized by androgen receptor (AR) signaling.
  • LAR cell lines were uniquely sensitive to bicalutamide (an AR antagonist).
  • TNBC GE profiles An extensive number of TNBC GE profiles was compiled with the intent of molecularly subtyping the disease. Six (6) TNBC subtypes were identified. Further, using GE signatures derived from each TNBC subtype, representative TNBC cell lines that serve as models for the different subtypes of the disease were aligned. Using the panel of cell lines, prominent signaling pathways revealed by GE signatures were pharmacologically targeted and it was found that the cell lines representing the various subtypes had different sensitivities to targeted therapies currently under laboratory and clinical investigation. The identification of diverse TNBC subtypes and the molecular drivers in corresponding cell line models provides great insight to the heterogeneity of this disease and provides preclinical platforms for the development of effective treatment (see Lehmann, B. D., et al., J Clin Invest, 121(7): 2750-2767 (July 2011), the entire teachings of which are incorporated herein by reference).
  • PIK3CA kinase mutations are a frequent event in the LAR subtype (65.3% vs. 38.4%) and were not just selected for in vitro during establishment of the tumor-derived cell lines.
  • PI3K pathway activation plays a role in resistance to AR antagonists, as there are elevated levels of activated AKT in residual tumors after bicalutamide treatment of xenograft tumors.
  • genetic or pharmacological targeting of AR synergizes with PI3K/mTOR inhibition in both two- and three-dimensional cell culture models.
  • triple negative breast cancer refers to breast cancers that are estrogen receptor (ER) negative, progesterone receptor (PR) negative and human epidermal growth factor receptor 2 (HER-2) negative.
  • ER estrogen receptor
  • PR progesterone receptor
  • HER-2 human epidermal growth factor receptor 2
  • the invention is based, in part, of the discovery of six TNBC subtypes.
  • TNBC triple negative breast cancer
  • methods of determining a triple negative breast cancer (TNBC) subtype in an individual in need thereof comprising determining expression of one or more genes (e.g., presence of one or more mRNAs and/or protein encoded by gene) in one or more TNBC cells of the individual; and comparing the expression of the one or more genes in the TNBC cells with the expression of the one or more genes in a control.
  • Increased expression of one or more genes comprising one or more cell cycle genes, cell division genes, proliferation genes, DNA damage response genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC BL1 subtype.
  • Increased expression of one or more growth factor signaling genes, glycolysis genes, gluconeogenesis genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC BL2 subtype.
  • Increased expression of one or more immune cell signaling genes, cytokine signaling genes, antigen processing and presentation genes, core immune signal transduction genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC IM subtype.
  • Increased expression of one or more cell motility genes, extracellular matrix (ECM) receptor interaction genes, cell differentiation genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC M subtype.
  • TNBC MSL subtype Increased expression of one or more hormone regulated genes in the TNBC cells compared to the control indicates that the TNBC is a TNBC LAR type.
  • the invention is directed to a method of determining a TNBC BL1 subtype in an individual in need thereof comprising determining increased expression of one or more cell cycle genes, cell division genes, proliferation genes, DNA damage response genes or a combination thereof in the TNBC cells of the individual compared to the control. Increased expression of the one or more genes indicates that the TNBC is a BL1 subtype TNBC.
  • the method of determining a TNBC BL1 subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising AURKA, AURKB, CENPA, CENPF, BUB1, BUB1B, TTK, CCNA2, PLK1, PRC1, MYC, NRAS, PLK1, BIRC5, CHEK1, FANCA, FANCG, FOXM1, HNGA1, RAD54BP, RAD51, NEKS, NBN, EXO1, MSH2, MCM10, RAD21,SIX3, Z1C1, SOX4, SOX1 and MDC1 wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC BL1 subtype.
  • a gene combination comprising AURKA, AURKB, CENPA, CENPF, BUB1, BUB1B, TTK, CCNA2, PLK1, PRC1, MYC, NRAS, PLK1, BIRC5, CHEK1, FANCA, FANCG, FOXM
  • Determination that the TNBC is a TNBC BL1 subtype can further comprise detecting increased expression of Ki-67 mRNA in the TNBC cells compared to the control.
  • determination that the TNBC is a TNBC BL1 subtype can further comprise detecting one or more mutated genes. Examples of such mutated genes include mutated BRCA1, STAT4, UTX, BRCA2, TP53, CTNND1, TOP2B, CAMK1G, MAPK13, MDC1, PTEN, RB1, SMAD4, CDKN2A, ATM, ATR, CLSPN, HDAC4, NOTCH1, SMARCAL1, and TIMELESS.
  • the invention is directed to a method of determining a TNBC BL2 subtype in an individual in need thereof comprising determining increased expression of one or more growth factor signaling genes, glycolysis genes, gluconeogenesis genes or a combination thereof in the TNBC cells compared to the control. Increased expression of the one or more genes indicates that the TNBC is a TNBC BL2 subtype.
  • the method of determining a TNBC BL subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising EGFR, MET, ELF4, MAF, NUAK1, JAG1, FOSL2, 1D1, ZIC1, SOX11, 1D3, FHL2, EPHA2, TP63 and MME wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC BL2 subtype.
  • Determination that the TNBC is a TNBC BL2 subtype can further comprise detecting one or more mutated genes. Examples of such genes comprise mutated BRCA1, RB1, TP53, PTEN, CDKN2A, UTX, BRAC2, PTCH1, PTCH2, and RET.
  • the invention is directed to a method of determining a TNBC M subtype in an individual in need thereof comprising determining increased expression of one or more cell motility genes, extracellular matrix (ECM) receptor interaction genes, cell differentiation genes or a combination thereof in the TNBC cells compared to the control. Increased expression of the one or more genes indicates that the TNBC is a TNBC M subtype.
  • ECM extracellular matrix
  • the method of determining a TNBC M subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising TGFB1L1, BGN, SMAD6, SMAD7, NOTCH1, TGFB1, TGFB2, TGFB3, TGFBR1, TGFBR2, TGFBR3, MMP2, ACTA2, SNAI2, SPARC, SMAD7, PDGFRA, TAGLN, TCF4, TWIST1, ZEB1, COL3A1, JAG1, EN1, MYLK, STK38L, CDH11, ETV5, IGF1R, FGFR1, FGFR2, FGFR3, TBX3, COL5A2, GNG11, ZEB2, CTNNB1, DKK2, DKK3, SFPR4, TCF4, TCF7L2, FZD4, CAV1, CAV2, CCND1 and CCND2 wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC M subtype.
  • Determination that the TNBC is a TNBC M subtype can further comprise detecting decreased E-cadherin (CDH1) expression compared to the control.
  • Determination that the TNBC is a TNBC M subtype can further comprise detecting one or more mutated genes. Examples of such mutated genes include mutated PTEN, RB1, TP53, PIK3CA, APC, BRAF, CTNNB1, FGFR1, GLI1, HRAS, KRAS, NOTCH1, and NOTCH4.
  • Determination that the TNBC is a TNBC M subtype can further comprise detecting chromosomal amplifications in KRAS, IGF1R or MET.
  • the invention is directed to a method of determining a TNBC MSL subtype in an individual in need thereof comprising determining increased expression of one or more cell motility genes, cellular differentiation genes, growth pathway genes, growth factor signaling genes, angiogenesis genes, immune signaling genes, stem cell genes, HOX genes, mesenchymal stem cell-specific marker genes or a combination thereof in the TNBC cells compared to the control. Increased expression of the one or more genes indicates that the TNBC is a TNBC MSL subtype.
  • the method of determining a TNBC MSL subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising VEGFR2 (KDR), TEK, TIE1, EPAS1, ABCA8, PROCR, ENG, ALDHA1, PER1, ABCB1, TERF2IP, BCL2, BMP2, EPAS1, STAT4, PPARG, JAK1, ID1, SMAD3, TWIST1, THY1, HOXA5, HOXA10, GL13, HHEX, ZFHX4, HMBOX1, FOS, PIK3R1, MAF, MAFB, RPS6KA2, TCF4, TGFB1L1, MEIS1, MEIS2, MEOX1, MEOX2, MSX1, ITGAV, KDR, NGFR, NTSE, PDGFRA, PDGFRB, POU2F1, and VCAM1 wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC MSL subtype.
  • VEGFR2 VEGFR2
  • Determination that the TNBC is a TNBC MSL subtype can further comprise detecting low expression of claudins 3, 4, 7 or a combination thereof compared to the control. Determination that the TNBC is a MSL subtype TNBC can further comprise detecting one or more mutated genes. Examples of such mutated genes include mutated CDKN2A, HRAS, TP53, NF1, PIK3CA, BRCA1, BRAF, KRAS, NF2, PDGFRA, APC, CTNNB1, FGFR1, PDGFRB.
  • the invention is directed to a method of determining a TNBC IM subtype in an individual in need thereof comprising determining increased expression of one or more immune cell signaling genes, cytokine signaling genes, antigen processing and presentation genes, core immune signal transduction genes or a combination thereof in the TNBC cells compared to the control. Increased expression of the one or more genes indicates that the TNBC is a TNBC IM subtype.
  • the method of determining a TNBC IM subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising CCL19, CCL3, CCL4, CCL5, CCL8, CCR1, CCR2, CCR5, CCR7, CD2, CD37, CD38, CD3D, CD48, CD52, CD69, CD74, and CD8A wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC IM subtype.
  • Determination that the TNBC is a TNBC IM subtype can further comprise detecting one or more mutated genes. Examples of such mutated genes include TP53, CTNNA1, DDX18, HUWE1, NFKBIA, APC, BRAF, MAP2K4, RB1, STAT4, STAT1, and RET.
  • the invention is directed to a method of determining a TNBC LAR subtype in an individual in need thereof comprising determining increased expression of one or more hormone regulated genes in the TNBC cells compared to the control. Increased expression of the one or more genes indicates that the TNBC is a TNBC LAR type.
  • the method of determining a TNBC LAR subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising AR, DHCR24, ALCAM, GATA2, GATA3, IDIH1, IDIH2, CDH11, ERBB3, CUX2, FGFR4, HOPX, FASN, FKBP5, APOD, PIP, SPDEF, CLDN8, FOXA1, KRT18, and XBP1 wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC LAR subtype.
  • Determination that the TNBC is a LAR subtype TNBC can further comprise detecting one or more mutated genes. Examples of such mutated genes include PIK3CA, CDH1, PTEN, RB1, TP53, and MAP3K1.
  • a variety of methods can be used to detect increased expression of a (one or more) gene.
  • increased expression of one or more genes can be detected by measuring all or a portion of nucleic acid (e.g., DNA, mRNA) of the gene and/or protein (polypeptide) expressed by the gene. The level, expression and/or activity of a gene and or its encoded polypeptide can be measured.
  • nucleic acid such as genes, mRNA or a combination thereof
  • PCR e.g., RT-PCR, RT-qPCR
  • gene chips or microarray analysis DNA arrays, gene expression arrays, nanostrings
  • next generation sequencing e.g., next generation RNA sequencing
  • in situ hybridization e.g., blotting techniques, and the like.
  • Protein can be detected using immunoassays (e.g., immunohistochemistry, immunofluorescence, immunoprecipitation), arrays (e.g., reverse phase protein microarrays), blotting techniques (e.g., Western blots), SDS-PAGE, and the like
  • the methods can further comprise detecting one or more mutated genes.
  • a variety of methods can be used to detect one or more mutated genes. Examples of such methods include a SNaPshot assay, exome sequencing, Sanger gene sequencing, resequencing array analysis, mRNA analysis/cDNA sequencing polymerase chain reaction (PCR), single-strand conformation polymorphism (sscp), heteroduplex analysis (het), allele-specific oligonucleotide (aso), restriction fragment analysis, allele-specific amplification (asa), single nucleotide primer extension, oligonucleotide ligation assay (ola), denaturing gradient gel electrophoresis (DGGE), temperature gradient gel electrophoresis (TGGE) and single strand conformation polymorphism (SSCP).
  • PCR mRNA analysis/cDNA sequencing polymerase chain reaction
  • sscp single-strand conformation polymorphism
  • heteroduplex analysis het
  • the methods can further comprise comparing the expression of the one or more genes in the individual to the expression of the one or more genes in a control.
  • a variety of suitable controls can be used. Examples of such controls include one or more non-cancer cells (e.g., breast cells from one or more individuals that do not have cancer) and/or non-TNBC cells (e.g., from the individual being tested).
  • the control can also be a (one or more) standard or reference control established by assaying one or more (e.g., a large sample of) individuals which do not have cancer (e.g., TNBC) and using a statistical model to obtain a control value (standard value; known standard). See, for example, models described in Knapp, R. G. and Miller M. C. (1992) Clinical Epidemiology and Biostatistics , William and Wilkins, Harual Publishing Co. Malvern, PA, which is incorporated herein by reference.
  • an “individual” refers to an animal, and in a particular aspect, a mammal.
  • mammals include primates, a canine, a feline, a rodent, and the like.
  • Specific examples include humans, dogs, cats, horses, cows, sheep, goats, rabbits, guinea pigs, rats and mice.
  • an individual in need thereof refers to an individual who is in need of treatment or prophylaxis as determined by a researcher, veterinarian, medical doctor or other clinician.
  • an individual in need thereof is a mammal, such as a human.
  • the individual has been diagnosed with breast cancer, the individual has not been diagnosed with breast cancer, or the individual is at risk of developing breast cancer.
  • the methods of the invention can further comprise detecting expression (increased expression) of one or more genes in a sample (biological sample) of the individual.
  • the method can further comprise obtaining a (one or more) sample from the individual.
  • the sample can be, for example, a biological fluid, a biological tissue (e.g., a biopsy) and combinations thereof from the individual.
  • the sample is one or more TNBC cells (e.g., epithelial cells, mesenchymal cells, immune cells). Methods for obtaining such biological samples from an individual are known to those of skill in the art.
  • the methods can further comprise determining a treatment protocol for the triple negative breast cancer (TNBC) patient based on the TNBC subtype.
  • a “cancer therapy”, “treatment for cancer” or “cancer treatment” comprises administering one or more agents to the individual.
  • the agent is a chemotherapeutic agent which targets the TNBC cells.
  • the agent is a targeted therapy agent which attacks cancer cells more specifically than chemotherapeutic agents.
  • agents include imatinib (Gleevec), gefitinib (Iressa), erlotinib (Tarceva), rituximab (Rittman), and bevacizumab (Avastin).
  • the agent is a sex hormone, or hormone-like drug that alters the action or production of female or male hormones and can be used to slow the growth of breast, prostate, and endometrial (uterine) cancers.
  • agents include anti-estrogens (e.g., tamoxifen, fulvestrant), aromatase inhibitors (e.g., anastrozole, exemestane, letrozole), progestins (e.g., megestrol acetate), anti-androgens (e.g., bicalutamide, flutamide) and LHRH agonists (leuprolide, goserelin).
  • the agent can be a drug which is used to stimulate the immune system to more effectively recognize and attack cancer cells of an individual with cancer.
  • agents include a hormone such as a selective estrogen receptor modulator (SERM); an antibody or antigen binding fragment thereof (e.g., herceptin); a protein tyrosine kinase inhibitor; a combination of chemotherapeutic agents such as cytoxan (C), methotrexate (M), fluorouracil (F), an anthracylcin such as adriamycin (A), epirubicin (E), doxorubicin, and/or daunorubicin; a targeted kinase inhibitor; a metalloproteinase inhibitor; and a proteosome inhibitor.
  • SERM selective estrogen receptor modulator
  • herceptin e.g., herceptin
  • F protein tyrosine kinase inhibitor
  • chemotherapeutic agents such as cytoxan (C), methotrexate (M), fluorouracil (F), an anthracylcin such as adriamycin (A), epirubicin
  • the one or more agents can be administered to the individual before (e.g., neoadjuvant therapy), during or after (e.g., adjuvant therapy) primary treatment of the cancer.
  • primary treatment of a cancer generally refers to a treatment involving surgery (e.g., surgical removal of a tumor) and/or radiation (e.g., local radiation).
  • the “treatment for cancer” can be a neoadjuvant treatment wherein an agent (e.g., a hormone, a chemotherapeutic) is administered prior to surgical removal of a (residual) tumor, or prior to localized radiation (e.g., radiation used to shrink the tumor in order to simplify subsequent surgical removal of the tumor).
  • an agent e.g., a hormone, a chemotherapeutic
  • the “treatment for cancer” can be an adjuvant treatment wherein an agent (e.g., a hormone, a chemotherapeutic agent) is administered after surgical removal of a (residual) tumor, or after localized radiation.
  • the treatment can be a palliative treatment in which one or more chemotherapeutic agents are administered to an individual to treat metastatic cancer (e.g., to make the individual more comfortable and/or to prolong survival of the individual after the cancer has metastasized).
  • the TNBC BL1 or BL2 subtype is detected in the individual and the individual is treated with one or more drugs that damages DNA.
  • drugs include an alkylating-like agent (e.g., cisplatin) and a PARP inhibitor (e.g., veliparib, olaparib).
  • the TNBC ML subtype is detected in the individual and the individual is treated with one or more drugs that inhibits Src (e.g., disatinib), IGF1R (e.g., OSI-906, BMS-754807), MET (e.g., PF2341066), PDGFR (e.g., sorafinib), P13K (e.g., BKM-120, GDC0941), P13K/mTOR (e.g., NVP-BEZ235, GDC0980) or a combination thereof.
  • Src e.g., disatinib
  • IGF1R e.g., OSI-906, BMS-754807
  • MET e.g., PF2341066
  • PDGFR e.g., sorafinib
  • P13K e.g., BKM-120, GDC0941
  • P13K/mTOR e.g., NVP-BEZ235, GDC09
  • the TNBC LAR subtype is detected in the individual and the individual is treated with one or more drugs that inhibit androgen receptor (AR).
  • AR androgen receptor
  • examples of such drugs include bicalutamide, MVD3100, and abiraterone.
  • the method can further comprise treating the individual with P13K/mTOR (e.g., NVP-BEZ235, GDC0980) or a combination thereof.
  • the method can further comprise treating the individual with an inhibitor of HSP90 (e.g., DMAG).
  • the invention can further comprise predicting whether an individual (e.g., patient) will benefit from a (one or more) treatment for a particular TNBC subtype.
  • the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) subtype comprising contacting a cell line that is a model for the TNBC subtype of interest with an agent to be assessed; and determining viability of the cell line in the presence of the agent, wherein if the viability of the cell line decreases in the presence of the agent, then the agent can be used to treat the TNBC subtype of interest.
  • TNBC triple negative breast cancer
  • the TNBC subtype of interest can be a TNBC BL-1 subtype, a TNBC BL-2 subtype, a TNBC IM subtype, a TNBC M subtype, a TNBC MSL subtype or a TNBC LAR subtype.
  • the viability of a cell decreases when e.g., the cell undergoes apoptosis, proliferation of the cell is inhibited or slowed and/or metastasis of the cell is inhibited to slowed.
  • test agent examples include nucleic acids, polypeptides, fusion proteins, peptidomimetics, prodrugs, drugs, receptors, binding agents (e.g., antibodies), small molecules, etc and libraries (e.g., combinatorial libraries) of such agents.
  • Test agents can be obtained made or obtained from libraries of natural, synthetic, and/or semi-synthetic products (e.g., extracts). Those skilled in the field of drug discovery and development will understand the precise source of such agents.
  • the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) BL-1 subtype comprising contacting one or more TNBC BL-1 subtype cell lines with an agent to be assessed.
  • TNBC triple negative breast cancer
  • the viability of the one or more cell lines in the presence of the agent is detected, wherein if the viability of the one or more cell lines decreases in the presence of the agent, then the agent can be used to treat the TNBC BL-1 subtype.
  • TNBC BL-1 subtype cell lines include HCC2157, HCC1599, HCC1937, HCC1143, HCC3153, MDA-MB-468, and HCC38.
  • the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) BL-2 subtype comprising contacting one or more TNBC BL-2 subtype cell lines with an agent to be assessed.
  • TNBC triple negative breast cancer
  • the viability of the one or more cell lines in the presence of the agent is determined, wherein if the viability of the one or more cell lines decreases in the presence of the agent, then the agent can be used to treat the TNBC BL-2 subtype.
  • TNBC BL-2 subtype cell lines include SUM149PT, CAL-851, HCC70, HCC1806 and HDQ-P1.
  • the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) IM subtype comprising contacting one or more TNBC IM subtype cell lines with an agent to be assessed.
  • TNBC triple negative breast cancer
  • the viability of the one or more cell lines in the presence of the agent is determined, wherein if the viability of the one or more cell lines decreases in the presence of the agent, then the agent can be used to treat the TNBC IM subtype.
  • TNBC IM subtype cell lines include HCC1187 and DU4475.
  • the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) M subtype comprising contacting one or more TNBC M subtype cell lines with an agent to be assessed.
  • TNBC M subtype cell lines include BT-549, CAL-51 and CAL-120.
  • the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) MSL subtype comprising contacting one or more TNBC MSL subtype cell lines with an agent to be assessed.
  • TNBC MSL subtype cell lines include HS578T, MDA-MB-157, SUM159PT, MDA-MB-436, and MDA-MB-231.
  • the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) LAR subtype comprising contacting one or more TNBC LAR subtype cell lines with an agent to be assessed.
  • the viability of the one or more cell lines in the presence of the agent is determined, wherein if the viability of the one or more cell lines decreases in the presence of the agent, then the agent can be used to treat the TNBC LAR subtype.
  • TNBC LAR subtype cell lines include MDA-MB, SUM185PE, HCC2185, CAL-148, and MFM-223.
  • viability of one of more of the cells can be determined using a variety of methods.
  • methods include a membrane leakage assay, a mitochondrial activity assay, a functional assay, a proteomic assay, a genomic assay or a combination thereof.
  • the methods can further comprise comparing the viability of the one or more cell lines in the presence of the agent to the viability of a control such as a non-cancerous cell line (e.g., a culture of normal human and immortalized MCF210A).
  • a control such as a non-cancerous cell line (e.g., a culture of normal human and immortalized MCF210A).
  • TNBC tumors were renormalized along with 5 positive controls for each parameter (ER, PR, and HER2) from 4 data sets that were positive by IHC and expressed mRNA near the center of the positive bimodal peak ( FIG. 1 B ). Only tumors that displayed a greater than 10-fold reduction in expression than the positive controls were considered negative in expression and used for further analysis, resulting in 386 TNBC tumors (training set) and 201 TNBC tumors (validation set).
  • STR Short tandem repeat
  • ICH Inhibitory concentration
  • mice Five-week-old female athymic nude-Foxnlnu mice (Harlan Sprague-Dawley) were injected (s.c.) with either approximately 5 ⁇ 10 6 (CAL-51, HCC1806, MDA-MB-468, and SUM185PE) or approximately 10 ⁇ 10 6 (CAL-148 and SUM159PT) cells suspended in medium (200 ⁇ l) into each flank using a 22-gauge needle.
  • the protocols describing the process of xenograft tumor formation in athymic mice were reviewed and approved by the Vanderbilt Institutional Review Board. Once tumors reached a volume of 25-50 mm 3 , mice were randomly allocated to treatment or vehicle arms.
  • Treatments included bicalutamide per oral (100 mg/kg/d), cisplatin i.p. (8 mg/kg/wk), or NVP-BEZ235 per oral (50 mg/kg/d) in 1:9 N-methylpyrolidone: PEG300. Tumor diameters were serially measured at the indicated times with digital calipers, and tumor volumes were calculated as width 2 ⁇ length/2. Data points reflect the means and SEM for 16 tumors per treatment.
  • FFPE tissue was subject to antigen retrieval with high pH buffer (pH 8.0) followed by overnight incubation with an AR (1:30) or Ki67 (1:75) antibody dilution overnight.
  • AR expression was scored as both the percentage of tumor cells with nuclear staining as well as the intensity of staining (scored as 0-3+).
  • An AR intensity score was calculated as follows: (AR intensity ⁇ 100)+% AR positive cells. For Ki67 the percentage of cells demonstrating nuclear staining at any intensity was recorded.
  • MFM-223, MDA-MB-453 and CAL-148 cells (3000 cells/well) were reverse-transfected with 1.25 pmole of siRNAs to AR [ONTARGET plus SMARTpool, cat #L-003400-00 (Dharmacon, Lafayette, CO)] or non-targeting control (ON-TARGETplus Non-targeting Pool cat #D-001810-10-05) with 0.25 ⁇ L Dharmafect #3 (MFM-223), or 0.25 ⁇ L Dharmafect #1 (MDAMB-453 and CAL-148). Colonies were stained and quantified 14 d following transfection using Cell Profiler 2.0 (Broad Institute, Cambridge, MA). Experiments were preformed in triplicate and error bars reflect standard deviation.
  • the 386 TNBC GE profiles of the training set were robust multiarray average (RMA) normalized, summarized, transformed, and corrected for “batch effect,” resulting in 13,060 identical probes representing unique genes across all platforms.
  • the triple-negative GE pattern is shown compared with 5 positive controls for each parameter (ER, PR, and HER2) from data sets that were confirmed IHC positive and expressed mRNA near the center of the positive bimodal peak ( FIG. 1 C ).
  • 4 GSE-7904, E-TABM-158, MDA133, GSE-22513, GSE-28821, and GSE-28796) included IHC data for all 3 markers, while others lacked information on ER, PR, or HER2 status (Table 1).
  • the IHC data provided were used to calculate false-positive rates for each study, defined as tumors that were predicted negative for ER, PR, or HER2 by bimodal filtering of mRNA expression, but were positive by IHC.
  • the overall false-positive rates were 1.7%, 1.7%, and 0.9% for ER, PR, and HER2, respectively, demonstrating that bimodal filtering of data sets by mRNA expression is a reliable method for identifying TNBC tumors from data sets lacking IHC information (Table 1).
  • the overall frequency of TNBC across the training data set was 16% and is consistent with the prevalence of TNBC previously reported in 2 other large studies performed on 3,744 cases (17%) ( 17 ) and 1,726 cases (16%) ( 18 ).
  • k-means and consensus clustering reveal 6 TNBC subtypes.
  • k-means clustering was performed on the most differentially expressed genes (SD >0.8).
  • SD >0.8 silhouette width
  • k-means clustering classified 337 of the 386 TNBC tumors into 6 stable clusters (s[i]>0) and 49 tumors into 1 unstable cluster (s[i]>0) ( FIG. 2 A ).
  • Sample classification robustness was analyzed by consensus clustering, which involves k-means clustering by resampling (1,000 iterations) randomly selected tumor profiles.
  • the consensus matrix is a visual representation of the proportion of times in which 2 samples are clustered together across the resampling iterations ( FIG. 2 B ). Groups of samples that frequently cluster with one another are pictorially represented by darker shades of red.
  • CDF consensus distribution function
  • the optimal number of clusters is 7, as defined by the consensus plots consistent with the k-means clustering (6 stable, 1 unstable).
  • Unsupervised dimension reduction by principal component analysis demonstrated fundamental differences in GE between tumor subtypes identified by k-means and consensus clustering ( FIG. 2 E ).
  • TNBC samples were compiled from bimodal filtering of 7 additional publicly available data sets (Table 2).
  • a GE signature was derived from the most differentially expressed genes found in the TNBC training set (see Methods) and used to predict which TNBC subtype was best-fit for each of the tumors in the validation set.
  • Each sample from the validation set was assigned to 1 of the TNBC subtypes derived from the training data set based on the highest Pearson correlation and lowest P value. Samples with correlations differing by P ⁇ 0.05 were considered unclassifiable.
  • the validation set resulted in proportioned subtypes similar to those of the initial k-means clustering of the training set.
  • GSE-A Gene Set Enrichment Analysis
  • TNBC subtypes were characterized on the basis of gene ontologies and differential GE and subsequently labeled as follows: basal-like 1 (BL1); basal-like 2 (BL2); immunomodulatory (IM); mesenchymal (M); mesenchymal stem-like (MSL); luminal androgen receptor (LAR); and unstable (UNS) ( FIG. 3 ).
  • BL1 basal-like 1
  • BL2 basal-like 2
  • IM immunomodulatory
  • M mesenchymal
  • MSL mesenchymal stem-like
  • LAR luminal androgen receptor
  • UMS unstable
  • BL1 and BL2 subtypes The top gene ontologies for the BL1 subtype are heavily enriched in cell cycle and cell division components and pathways (cell cycle, DNA replication reactome, G 2 cell-cycle pathway, RNA polymerase, and G 1 to S cell cycle) ( FIG. 3 ).
  • the annotations are supported by the expression of genes associated with proliferation, such as AURKA, AURKB, CENPA, CENPF, BUB1, TTK, CCNA2, PRC1, MYC, NRAS, PLK1, and BIRC5 ( FIG. 13 ). Elevated DNA damage response (ATR/BRCA) pathways accompany the proliferation pathways in the BL1 subtype ( FIG. 3 ).
  • the BL2 subtype displays unique gene ontologies involving growth factor signaling (EGF pathway, NGF pathway, MET pathway, Wnt/ ⁇ -catenin, and IGF1R pathway) as well as glycolysis and gluconeogenesis ( FIG. 3 ).
  • the BL2 subtype is uniquely enriched in growth factor receptor GE such as EGFR, MET, and EPHA2 ( FIG. 13 ).
  • This subtype has features suggestive of basal/myoepithelial origin as demonstrated by higher expression levels of TP63 and MME (CD10) ( FIG. 13 ).
  • the IM subtype is enriched for gene ontologies in immune cell processes. These processes include immune cell signaling (TH1/TH2 pathway, NK cell pathway, B cell receptor [BCR] signaling pathway, DC pathway, and T cell receptor signaling), cytokine signaling (cytokine pathway, IL-12 pathway, and IL-7 pathway), antigen processing and presentation, and signaling through core immune signal transduction pathways (NFKB, TNF, and JAK/STAT signaling) ( FIG. 3 ).
  • the IM signaling is evidenced by immune signal transduction GE ( FIG. 13 ), in addition to immune cell-surface antigens, cytokine signaling, complement cascade, chemokine receptors and ligands, and antigen presentation ( FIG. 16 ).
  • Immune signaling genes within the IM subtype substantially overlap with a gene signature for medullary breast cancer, a rare, histologically distinct form of TNBC that despite its high-grade histology is associated with a favorable prognosis (Bertucci F, et al. Cancer Res. 2006; 66(9):4636-4644).
  • the M subtype displays a variety of unique gene ontologies that are heavily enriched in components and pathways involved in cell motility (regulation of actin by Rho), ECM receptor interaction, and cell differentiation pathways (Wnt pathway, anaplastic lymphoma kinase [ALK] pathway, and TGF- ⁇ signaling) ( FIG. 3 ).
  • the MSL subtype shares enrichment of genes for similar biological processes with the M subtype, including cell motility (Rho pathway), cellular differentiation, and growth pathways (ALK pathway, TGF- ⁇ signaling and Wnt/ ⁇ -catenin pathway).
  • genes representing components and processes linked to growth factor signaling pathways that include inositol phosphate metabolism, EGFR, PDGF, calcium signaling, G-protein coupled receptor, and ERK1/2 signaling as well as ABC transporter and adipocytokine signaling ( FIG. 3 ).
  • TGF- ⁇ signaling pathway components TGFB1L1, BGN SMAD6, SMAD7, NOTCH1, TGFB1, TGFB2, TGFB3, TGFBR1, TGFBR2, and TGFBR3
  • EMT-associated epithelial-mesenchymal transition-associated genes
  • growth factors FGF, IGF, and PDGF pathways
  • Wnt/ ⁇ -catenin signaling CNNB1, DKK2, DKK3, SFRP4, TCF4, TCF7L2, FZD4, CAV1, CAV2, and CCND2
  • the MSL subtype is also uniquely enriched in genes involved in angiogenesis, including VEGFR2 (KDR), TEK, TIE1, and EPAS1 as well as immune signaling evidenced by an overlap in GE unique to the IM subtype ( FIG. 16 ).
  • MSL subtype expresses low levels of proliferation genes ( FIG. 13 ). This decreased proliferation is accompanied by enrichment in the expression of genes associated with stem cells (ABCA8, PROCR, ENG, ALDHA1, PER1, ABCB1, TERF2IP BCL2, BMP2, and THY1), numerous HOX genes (HOXA5, HOXA10, MEIS1, MEIS2, MEOX1, MEOX2, and MSX1), and mesenchymal stem cell-specific markers (BMP2, ENG, ITGAV, KDR, NGFR, NT5E, PDGFRB, THY1, and VCAM1) ( FIG. 13 ).
  • the signaling pathway components differentially expressed in the M and MSL groups share similar features with a highly dedifferentiated type of breast cancer called metaplastic breast cancer, which is characterized by mesenchymal/sarcomatoid or squamous features and is chemoresistant (Gibson G R, et al. Am Surg. 2005; 71(9):725-730).
  • the MSL subtype also displays low expression of claudins 3, 4, and 7, consistent with a recently identified claudin-low subtype of breast cancer ( FIGS. 13 and 17 ; Prat et al., Breast Cancer Res, 2010; 12(5):R68).
  • This population of claudin-low-expressing tumors also had high expression of genes associated with EMT (FBN1, MMP2, PDGFRB, THY1, SPARC, TGFBR2, PDGFRA, TWIST, CAV1, CAV2, and SERPINE1).
  • LAR subtype GE in the LAR group was the most differential among TNBC subtypes. This subtype is ER negative, but gene ontologies are heavily enriched in hormonally regulated pathways including steroid synthesis, porphyrin metabolism, and androgen/estrogen metabolism ( FIG. 3 ). Whether other hormone-regulated pathways such as androgen receptor (AR) signaling, previously reported in ER-negative breast cancer (Hayes M J et al. Clin Cancer Res. 2008; 14(13):4038-4044), could be responsible for the GE patterns in this LAR subtype was investigated. Indeed, it was found that AR mRNA was highly expressed, on average at 9-fold greater than all other subtypes ( FIG. 13 ).
  • AR androgen receptor
  • Tumors within the LAR group also expressed numerous downstream AR targets and coactivators (DHCR24, ALCAM, FASN, FKBP5, APOD, PIP, SPDEF, and CLDN8) ( FIG. 13 ).
  • Hierarchical clustering was performed using an AR-activated gene signature (Chen C D, et al. Nat Med.
  • FIGS. 19 A- 19 B Tumors in the LAR subtype display luminal GE patterns, with FOXA1, KRT18, and XBP1 among the most highly expressed genes ( FIG. 20 A ).
  • Others have previously described a breast cancer subgroup expressing AR termed molecular apocrine (Chen C D, et al. Nat Med. 2004; 10(1):33-39).
  • the TNBC subgroup GE centroids were used to correlate the apocrine samples from GSE-1561 (Farmer P, et al. Oncogene. 2005; 24(29):4660-4671; Table 1).
  • the GE profiles of all 6 apocrine tumors (GSM26883, GSM26878, GSM26886, GSM26887, GSM26903, and GSM26910) described in the study strongly correlate with LAR, suggesting that the LAR TNBC subtype is composed of AR-driven tumors that include the molecular apocrine subtype.
  • Intrinsic molecular breast cancer subtype classification of TNBC Breast cancers can be classified as luminal or basal-like, dependent on their expression of different cytokeratins.
  • TNBC tumor subtypes display differential expression of both basal-like cytokeratins (KRT5, KRT6A, KRT6B, KRT14, KRT16, KRT17, KRT23, and KRT81) and luminal cytokeratins (KRT7, KRT8, KRT18, and KRT19) across the subtypes ( FIGS. 20 A- 20 B ).
  • the UNS, BL1, BL2, and M subtypes expressed higher levels of basal cytokeratin expression, while tumors within the LAR subtype lacked basal cytokeratin expression and expressed high levels of luminal cytokeratins and other luminal markers (FOXA1 and XBP1) ( FIG. 20 A ).
  • breast cancers can be classified by an “intrinsic/UNC” 306-gene set into 5 major subtypes (basal-like, HER2-like, normal breast-like, luminal A, and luminal B) (Hu Z, et al. BMC Genomics. 2006; 7:96).
  • each of the 386 TNBC tumor profiles were correlated to the intrinsic gene set centroids of the 5 molecular subtypes, as previously described (Hu Z, et al. BMC Genomics. 2006; 7:96). Tumors were assigned to 1 of the molecular subtypes based on the highest correlation coefficient between each TNBC expression profile and the 5 molecular subtype centroids.
  • the BL2, IM, and M subtypes only moderately correlated to the basal-like molecular class (31%, 58%, and 47%, respectively), with a portion of tumors unclassified (22%, 17%, and 18%, respectively) ( FIG. 20 B ).
  • the M and MSL subtypes displayed the largest portion of tumors classified as normal breast-like (25%, and 46%, respectively).
  • the BL2, M, and MSL subtypes were a mixture of classifications, suggesting the intrinsic classification system may not be suitable for characterizing these TNBC subtypes.
  • the majority of TNBC tumors within the LAR subtype were classified as either luminal A or luminal B (82%), and none were classified as basal-like, further supporting the luminal origin of the LAR subtype ( FIGS.
  • Relapse-free survival and distant-metastasis-free survival differs among TNBC subtypes.
  • DMFS Distant-metastasis-free survival
  • TNBC cell line models for TNBC subtypes are TNBC subtypes.
  • Cell line models would facilitate preclinical experiments to define differential drug sensitivity of the distinct subtypes within this heterogeneous disease.
  • ER, PR, and HER2 GE levels from 2 independent breast cancer cell line data sets (GSE-10890 and ETABM-157)
  • 24 and 25 triple-negative cell lines were identified in the GSE-10890 and ETABM-157 GE data sets, respectively.
  • Discrepancies in some cell lines e.g., HCC1500 and HCC1937
  • HCC1500 and HCC1937 may be the result of differences in culturing methods and/or loss of hormone receptor expression over time in culture. Analysis of these 2 data sets identified 30 nonoverlapping TNBC cell lines.
  • GE profiles from the cell lines were correlated to the centroids for each of the 6 TNBC subtypes.
  • the majority of cell lines (27 of 30) were assigned to TNBC subtypes (Table 3), except for BT20, HCC1395, and SW527, which had low correlations ( ⁇ 0.1) or were similar between multiple subtypes (P >0.05).
  • 7 cell lines that contained known BRCA1 and BRCA2 mutations 5 correlated with the BL1 or BL2 subtypes (HCC1937, HCC1599, HCC2157, HCC3153, and SUM149PT), consistent with this subtype containing tumors with defects in DNA repair pathways (Table 3) and gene ontologies enriched for GE involved in cell cycle and DNA repair functions ( FIG. 13 ).
  • Cell lines in the M and MSL subtypes were generated from highly dedifferentiated tumors derived from unique pathologies (e.g., HS578T, carcinosarcoma; and SUM159PT, anaplastic carcinoma) and expressed both epithelial and mesenchymal components. All cell lines assigned to the M and MSL subtype have spindle-like morphology in 2D culture (CAL-120, CAL-51, MDA-MB-157, MDA-MB-231, MDA-MB-436, SUM159PT, HS578T, and BT549) or stellate-like morphology in 3D (Kenny P A et al., Mol Oncol, 2007; 1:84-96). Five cell lines matched to the LAR subtype (MDA-MB-453, SUM185PE, HCC2185, CAL-148, and MFM-223).
  • basal A cell lines display epithelial characteristics and are associated with BRCA1 gene signatures, while basal B cell lines are more invasive and display mesenchymal and stem/progenitor-like characteristics.
  • the GE analyses revealed that the majority of basal A cell lines belong to the BL1 and BL2 subtypes, while the majority of basal B cell lines fall into the M and MSL subtypes (Table 3).
  • Hierarchical clustering analysis was tested on all TNBC cell lines using the most differentially expressed genes from the tumors to determine whether GE patterns of the cell lines are similar within TNBC subtypes ( FIG. 24 ). Three clusters were identified: LAR, containing all 4 LAR lines; basal-like, containing lines in the BL1 and BL2 subtypes; and mesenchymal-like, containing lines in the M and MSL subtypes. This clustering analysis indicates that TNBC cell lines can be classified into 3 main groups: basal-like (BL1 and BL2); mesenchymal-like (M and MSL); and LAR. This classification will be used in subsequent sections.
  • TNBC cell lines have differential sensitivity to therapeutic agents.
  • therapies undergoing clinical investigation in patients with TNBC including those targeting PARP (Farmer H, et al. Nature. 2005; 434(7035):917-921), AR (Agoff N S et al. Am J Clin Pathol. 2003; 120(5):725-731), Src (Finn R S, et al. Breast Cancer Res Treat. 2007; 105(3):319-326), and PI3K/mTOR (Marty B, et al. Breast Cancer Res. 2008; 10(6):R101) signaling.
  • the panel of TNBC cell lines was used to assess differential response to several agents targeting these pathways.
  • HMECs primary human mammary epithelial cells
  • IC 50 half-maximal inhibitory concentration
  • TNBC cell line panel was treated with the PARP inhibitors veliparib and olaparib, but not all cell lines representative of the basal-like TNBC subtypes were sensitive to PARP inhibition ( FIGS. 4 A- 4 D ).
  • the BRCA2-mutant cell line HCC1599 lacked sensitivity to either PARP inhibitor (veliparib IC 50 >30 ⁇ M and olaparib IC 50 >100 ⁇ M).
  • other properties of the tumor may dictate sensitivity to a given PARP inhibitor.
  • the BRCA1-mutant cells SUM149PT, HCC1937, and MDA-MB-436) and BRCA2-mutant cells (HCC1599) were among the most sensitive to cisplatin treatment ( FIG. 4 E ).
  • GE analysis of the mesenchymal-like subtypes demonstrated enrichment in the expression of genes that make up components and pathways associated with EMT (TGF ⁇ , ECM-receptor interaction, ALK, Wnt/ ⁇ -catenin, and Rac1) and those associated with cell motility (focal adhesion, integrin signaling, Rac1, striated muscle contraction, and regulation of actin by Rho GTPase) FIG. 3 ). Since the nonreceptor tyrosine kinase Src plays critical roles in cell migration and the mesenchymal-like subtypes are enriched in cell motility pathways, the effect of the Src inhibitor dasatinib on the panel of TNBC lines was analyzed.
  • TNBC cell lines Since activating mutations in PIK3CA are the most frequent genetic event in breast cancer (Samuels Y, et al. Science. 2004; 304(5670):554), the TNBC cell lines were treated with the dual PI3K/mTOR inhibitor NVP-BEZ235 (Maira S M, et al. Mol Cancer Ther. 2008; 7(7):1851-1863). TNBC cell lines that have activated PI3K/AKT signaling due to PIK3CA mutations or PTEN deficiency (Table 3) were highly sensitive to NVPBEZ235 ( FIG. 6 C ).
  • PIK3CA mutations frequent in the LAR subtype, with all LAR cell lines containing PIK3CA-activating mutations (HCC2185, MDA-MB-453 CAL-148, MFM-223, and SUM185PE) (Table 3). While PIK3CA mutations predicted NVP-BEZ235 sensitivity, PTEN deficiencies (mutation or loss of protein expression) did not correlate with sensitivity.
  • Xenograft tumors derived from TNBC cell lines display differential sensitivity to therapeutic agents in vivo.
  • xenograft tumors were established in nude mice from cell lines representative of the basal like (HCC1806 and MDA-MB-468), mesenchymal-like (CAL-51 and SUM159PT), or LAR (SUM185PE and CAL-148) subtypes. After tumors reached an approximate volume of 25-50 mm 3 , the mice were treated with cisplatin, NVP-BEZ235 or bicalutamide.
  • the sensitivity of the TNBC cell lines to the therapeutic agents when grown as 3D xenograft tumors in vivo was very similar to that seen with the cell lines grown in 2D monolayer culture.
  • the xenograft tumors derived from the 2 cell lines representative of basal-like tumors (HCC1806 and MDA-MB-468) were highly and differentially sensitive to cisplatin and were significantly growth inhibited (P ⁇ 0.0001) relative to treatment with vehicle control or the other experimental treatments (bicalutamide and NVP-BEZ235) ( FIG. 7 ).
  • the tumors derived from the LAR cell lines (SUM185PE and CAL-148) and the mesenchymal-like cell line that expresses low level AR protein (CAL-51) FIGS.
  • cell lines representative of the 6 TNBC subtypes display different sensitivities to a variety of agents, and importantly, these differences can be attributed to distinct expression of cellular components and presence of mutations in key oncogenes and tumor suppressors.
  • the results have immediate clinical translation, as they provide a valuable platform and insights for ongoing and future clinical investigation.
  • the data indicate that patients with basal-like TNBC should be treated with agents that engage DNA damage signaling response pathways; those with tumors expressing AR should receive bicalutamide alone or in combination with PI3K inhibitors; and those with mesenchymallike TNBC should be considered for trials exploring the activities of an Src antagonist in combination with a PI3K inhibitor.
  • TNBCs can be reliably identified by filtering GE profiles for ER, PR, and HER2 mRNA levels.
  • k-means and consensus clustering of tumor profiles revealed that TNBC is composed of 6 stable subtypes enriched in distinct gene ontologies and GE patterns.
  • cell-line models for each of the TNBC subtypes were identified.
  • TNBCs have been classified as basal-like either by IHC or by correlation to the intrinsic molecular breast cancer subtypes (Bertucci F, et al. Int J Cancer. 2008; 123(1):236-240; Rakha E A, et al. Clin Cancer Res. 2009; 15(7):2302-2310; Kreike B, et al. Breast Cancer Res. 2007; 9(5):R65).
  • a previous TNBC study identified 5 distinct hierarchical clusters in which 91% (88 of 97) of TNBCs identified by IHC correlated to the basal-like subtype (Kreike B, et al. Breast Cancer Res. 2007; 9(5):R65).
  • TNBC is not limited to tumors with a basal-like phenotype; rather it is a heterogeneous collection of tumors with distinct phenotypes, as evidenced by the diverse GE patterns and varying sensitivity of representative cell lines to the targeted therapies assessed in this study.
  • BL1 and BL2 subtypes express high levels of genes involved in cell proliferation and DNA damage response, suggesting patients with basal-like tumors would benefit from agents that preferentially target highly proliferative tumors (e.g., anti-mitotic and DNA-damaging agents). Consistent with this notion, patients with basal-like tumors had up to a 4-fold higher pCR after taxane-based and radiation-based treatment as compared with patients with tumors that displayed characteristics of the ML or LAR subtypes (Bauer J A, et al. Clin Cancer Res. 2010; 16(2):681-690; Juul N, et al. Lancet Oncol. 2010; 11(4):358-365).
  • BRCA1 is a relatively large protein (1863 aa) that forms numerous complexes that may not be entirely disrupted when BRCA1 and BRCA2 are mutated, as opposed to BRCA-null cells.
  • these drugs have been proven to be highly effective in the clinical trial setting (41% objective response rate; 6.2 month progression-free survival) (Audeh M W, et al. J Clin Oncol. 2009; 27:5500).
  • cisplatin as a “targeted” agent alone or in combination with antimitotics (taxanes) and/or radiation may benefit patients with this subtype and a current trial is underway with these agents (Mayer I A, et al. J Clin Oncol. 2010; 28(1):15).
  • the IM subtype is highly enriched in immune cell signaling.
  • Other studies have described the presence of immune response gene signatures in ER-negative and medullary breast cancers (Bertucci F, et al. Cancer Res. 2006; 66(9):4636-4644; Teschendorff A E et al. Genome Biol. 2007; 8(8):R157). Similar to these studies, it was found elevated expression of T cell—associated genes, immune transcription factors, IFN regulatory factors, TNF, complement pathway, and antigen processing. It cannot be ruled out that the GE profile of the IM subtype comprises, at least in part, stromal components including immune cell infiltrate. However, the finding that the same proportion of microdissected tumors belongs to this group argues against stromal contamination.
  • the M and MSL subtypes share similar gene ontologies and GE profiles involving TGF- ⁇ , mTOR, Rac1/Rho, Wnt/ ⁇ -catenin, FGFR, PDGFR, and VEGF signaling pathways. These signaling pathways are prominent in processes of EMT and stem cell-like properties of a CD44+CD24-population of normal mammary cells (Mani S A, et al. Cell. 2008; 133(4):704-715; Shipitsin M, et al. Cancer Cell. 2007; 11(3):259-273).
  • the MSL subtype is made up at least in part by the recently described claudin-low tumors, which lack luminal differentiation markers, high enrichment for EMT markers, immune response genes, and cancer stem cell-like features (Prat A, et al. Breast Cancer Res. 2010; 12(5):R68).
  • the M and MSL subtypes differed clinically, with patients in the M subtype presenting with shorter RFS. This may be a reflection of differences in proliferation, as the M subtype displayed higher expression of proliferation-associated genes, including Ki-67.
  • patients with the M and MSL subtypes had decreased 5-year DMFS, consistent with enrichment in pathways associated with metastasis and motility.
  • Tumors within the mesenchymal-like subtypes have GE profiles that are similar to those from mesenchymal cells and metaplastic breast cancers (Hennessy B T, et al. Cancer Res. 2009; 69(10):4116-4124).
  • Metaplastic breast cancers have lineage plasticity, including foci of spindle cell, as well as osseous or cartilaginous differentiation (Gibson G R, et al. Am Surg. 2005; 71(9):725-730).
  • a recent study found that 47% of metaplastic breast cancers sequenced have PIK3CA mutations and have higher phosho-AKT expression (Hennessy B T, et al. Cancer Res. 2009; 69(10):4116-4124).
  • TNBC mesenchymallike cell lines preferentially responded to the dual PI3K/mTOR inhibitor NVP-BEZ235.
  • This response to NVP-BEZ235 was demonstrated in cell lines that carry PIK3CA mutations in xenografts in addition to an MSL cell line (SUM159PT) that lacks PIK3CA mutation or PTEN deficiencies, suggesting that the PI3K/mTOR pathway is important in the mesenchymal-like subtype.
  • the mesenchymal-like subtypes were enriched in pathways associated with EMT and cell motility. There is evidence of a prominent role for Src in tumor cells that are highly invasive, such as those that have undergone EMT (Guarino M. J Cell Physiol. 2010; 223(1):14-26). Accordingly, we found that mesenchymal-like TNBC cell lines had differential sensitivity to dasatinib. Markers of EMT may have clinical value for patient preselection for trials using dasatinib. In addition, Wnt-signaling pathways regulate EMT and may contribute to tumor cell invasion (Shin S Y, et al. Cancer Res. 2010; 70(17):6715-6724).
  • the LAR subtype was readily subclassified by an AR gene signature and high levels of luminal cytokeratin expression.
  • GE analysis of the LAR subtype is consistent with a prior report of a subset of ER-negative tumors expressing AR-regulated genes (Doane A S, et al. Oncogene. 2006; 25(28):3994-4008).
  • Farmer et al. described an apocrine tumor subtype based on GE profiling that was characterized by AR expression distinguishing this tumor subtype from other basal-like tumors (Farmer P, et al. Oncogene. 2005; 24(29):4660-4671).
  • the prevalence of the LAR tumors was 11% (62 of 587) of TNBCs or 2% (62 of 3247) of all breast cancers.
  • Analysis of clinical data demonstrated that patients in the LAR subtype had higher RFS but no difference in DMFS compared with all other TNBC subtypes, suggesting these patients have local relapse.
  • the higher RFS could imply that this group of patients received ineffective therapies (standard chemotherapy); however, patients in the LAR group were significantly older at diagnosis and the extent of disease or age-associated comorbidities that affect the ability to deliver treatment as planned may have contributed to relapse.
  • LAR cell lines were also sensitive to PI3K inhibition. This sensitivity correlated with PIK3CA mutations. All 5 LAR cells lines have activating PIK3CA mutations and are sensitive to the PI3K inhibitor NVP-BEZ235, similar to ER-positive breast cancer in which PIK3CA mutations are common (Gonzalez-Angulo A M, et al. Clin Cancer Res. 2009; 15(7):2472-2478; Stemke-Hale K, et al. Cancer Res. 2008; 68(15):6084-6091).
  • the GE analysis of TNBC described herein demonstrates that with sufficient sample size, distinct subtypes of TNBC can be identified with putative molecular targets.
  • the analyses provides biomarkers that can be used for patient selection in the design of clinical trials for TNBC as well as identification of potential markers of response to treatment.
  • the identification of cell lines representing TNBC tumor subtypes provides key models for preclinical studies with newly developed targeted agents.
  • PIK3CA kinase mutations were a frequent event in AR-positive TNBC tumors. It was found that genetic or pharmacological targeting of AR in LAR cells increased the therapeutic benefit of PI3K/mTOR inhibition. Further, it was discovered that after bicalutamide treatment of mice bearing LAR xenograft tumors, the PI3K pathway signaling increased.
  • AR-positive TNBC tumors are enriched for PIK3CA kinase domain mutations.
  • High frequency PIK3CA mutations in AR-positive TNBC cell lines are described in Example 1.
  • Sanger sequencing was performed on 26 AR-positive TNBC cases. PCR-amplified regions from exons 9 and 20 that harbor the most frequently occurring activating mutations in PIK3CA were sequenced. Consistent with the observations in cell lines, nearly all of the detected mutations occurred at amino acid H1047 (20 of 21) with only one occurring at E545K.
  • RNA-seq data confirmed significant levels of AR RNA (6.8 vs. 0.3, p ⁇ 0.0001) and protein (1.7 vs. 0.35, p ⁇ 0.0001) in the LAR subtype.
  • PIK3CA mutations were relatively rare in TNBC cases in the TCGA dataset (4 of 66, 6.0%).
  • LAR TNBC Cell Lines have Increased PI3K Pathway Activation and are Sensitive to PI3K and PI3K/mTOR Inhibition.
  • RPPA was used to investigate AR, PTEN, active AKT (pS473 and PT308) and active GSK3 ⁇ (pS21/pS9) protein levels in LAR cell lines to determine if the PI3K pathway was active in AR expressing, PI3K mutant TNBC.
  • Activated PI3K signaling correlated with PIK3CA mutation and PTEN loss.
  • AR receptor protein in the LAR TNBC cell lines was expressed at levels equal to or greater than LNCaP cells.
  • AR-expressing cell lines displayed higher levels of phosphorylated AKT (S473) and phosphorylated ribosomal S6, confirming an activated PI3K pathway and downstream mTOR activation, respectively ( FIG. 27 ).
  • TNBC cell lines To determine the sensitivity of PIK3CA mutation-bearing LAR cell lines to PI3K inhibitors relative to other TNBC cell lines, a large panel of TNBC cell lines was treated with pan-PI3K inhibitors BKM120 (Novartis) and GDC-0941 (Genentech) or with the dual PI3K/mTOR inhibitors NVP-BEZ235 (Novartis) and GDC-0980 (Genentech). AR-expressing and other TNBC cell lines containing PIK3CA mutations were among the most sensitive to PI3K inhibition, as indicated by low half maximal inhibitory concentration (IC50) values ( FIGS. 28 A- 28 C- 2 ).
  • IC50 half maximal inhibitory concentration
  • FIGS. 29 A- 29 B Immunoblot analysis confirmed decreased AR protein levels in three AR-expressing TNBC cell lines infected with lentiviral particles containing non-targeting shRNA or two AR-targeting hairpins ( FIG. 29 A ). Both shRNA expression vectors targeting AR decreased cell viability across multiple doses of PI3K or PI3K/mTOR inhibitors ( FIG. 29 B ). Efficacy of AR-targeting in combination with PI3K or PI3K/mTOR inhibitors was independent of compound, as similar results were obtained with BKM-120 and NVP-BEZ235 ( FIG. 34 ).
  • FIGS. 31 A- 31 B activated caspase 3/7 levels after drug treatments were measured.
  • Cells were treated with a high dose of adriamycin (ADR, 3 ⁇ M) to measure the ability of cells to engage the apoptotic pathway.
  • ADR adriamycin
  • PI3K is Activated after Pharmacological Inhibition of AR.
  • mice were either treated with vehicle, bicalutamide, NVP-BEZ235, GDC-0980 alone or either NVP-BEZ235 or GDC-0980 in combination with bicalutamide. While each single agent alone significantly decreased tumor growth, both PI3K/mTOR inhibitors in combination with bicalutamide inhibited tumor growth to the greatest extent ( FIG. 32 C ). Thus, the combined inhibition of AR and PI3K appears to be a rational treatment strategy for AR-expressing TNBC and should be considered in future clinical trial design.
  • a 588 bp fragment was PCR-amplified from exon 20 of PIK3CA using the following amplification primers; foreword: TGACATTTGAGCAAAGACCTG (SEQ ID NO: 1), reverse: CATAACATGAAATTGCGCATT (SEQ ID NO: 2). Sanger sequencing was performed on the PCR amplicon using the following internal sequencing primers; Exon Forward: ACATCATTTGCTCCAAACTGA (SEQ ID NO: 3) Reverse: CCTATGCAATCGGTCTTTGC (SEQ ID NO: 4) (Samuels et al., Science, 304:554 (2004)).
  • the TaqMan PCR reaction mixture was assembled from a 2 ⁇ ddPCR Mastermix (Bio-Rad), 20 ⁇ primer, and probes (final concentrations of 900 and 250 nM, respectively) and template (variable volume) in a final volume of 20 ⁇ L.
  • Each assembled ddPCR reaction mixture was then loaded into the sample well of an eight-channel disposable droplet generator cartridge (Bio-Rad).
  • a volume of 70 ⁇ L of droplet generation oil (Bio-Rad) was loaded into the oil well for each channel.
  • the cartridge was placed into the droplet generator (Bio-Rad).
  • the cartridge was removed from the droplet generator, where the droplets that collected in the droplet well were then manually transferred with a multichannel pipet to a 96-well PCR plate.
  • the plate was heat-sealed with a foil seal and then placed on a conventional thermal cycler and amplified to the end-point (40 cycles). Thermal cycling conditions were 95° C. ⁇ 10 min (1 cycle), 94° C. ⁇ 30 s and 57° C. ⁇ 60 s (40 cycles), and 12° C. hold.
  • the 96-well PCR plate was loaded on the droplet reader (Bio-Rad), which automatically reads the droplets from each well of the plate. Analysis of the ddPCR data was performed with QuantaSoft analysis software (Bio-Rad) that accompanied the droplet reader.
  • Primer sequences Forward: 5′GATAAAACTGAGCAAGAGGCTTTGG′ (SEQ ID NO: 5), Reverse: 5′GCTGTTTAATTGTGTGGAAGATCCAA′ (SEQ ID NO: 6), Wild type probe: 5′-VIC-CCACCATGATGTGCA-MGB-3 (SEQ ID NO: 7), ‘H1047L probe:5′-FAM-CCACCATGATGAGCA-MGB-3′ (SEQ ID NO: 8), H1047R probe: 5′-FAM-CCACCATGATGCGCA-MGB-3′ (SEQ ID NO: 9).
  • RNA-seq (lvl 3), somatic mutations (lvl2) and reverse phase protein array (RPPA, lvl3) data for breast cancer was downloaded from The Cancer Genome Atlas (tcga-data.nci.nih.gov/).
  • RNA-seq data (RPKM) was combined and molecularly subtyped using the online TNBCtype software according to published methods (Chen et al., Cancer Informatics, 11:147-1556 (2012)). After removal of potential ER positive, 137 TNBC samples were identified and assigned a molecular subtype.
  • Tumor or cell lysates were two-fold-serial diluted for 5 dilutions (from undiluted to 1:16 dilution) and arrayed on nitrocellulose-coated slide in 11 ⁇ 11 format.
  • Samples were probed with antibodies by CSA amplification approach and visualized by DAB colorimetric reaction. Slides were scanned on a flatbed scanner to produce 16-bit tiff image and the density was quantified by MicroVigene.
  • Relative protein levels for each sample were determined by interpolation of each dilution curves from the standard curve (supercurve) of the slide (antibody) as previously described (Zhang et al., Bioinformatics, 25:650-654 (2009)). These values were Log 2 transformed and normalized for protein loading by transformation a to linear value. Linear normalized data was then median-centered for heatmap comparisons.
  • Viability was determined from measuring fluorescent intensity after metabolic reduction of Alamar Blue in the presence/absence of drug after 72 hours. Viability assays were performed in triplicate and replicates were normalized to untreated wells. Inhibitory concentration (IC 50 ) values were determined after double-log transformation of dose response curves as previously described (Lehmann et al., J Clin Invest, 121:2750-2767 (2011)).
  • 293FT cells were transfected with Lipofectamine 2000 (Invitrogen, Grand Island, NY) the packaging vectors PAX2 and pMD2.g (Addgene, Cambridge, MA) along with pLKO.1-puro Misson shRNA constructs (Sigma) targeting AR or a non-targeting control.
  • Viral media was harvested 48 h post-transfection and added to target cells along with polybrene (10 ⁇ g/mL).
  • 72 h post-infection MDA-MB-453 8,000 cells/well
  • CAL-148 5,000 cells/well
  • MFM-223 7,000 cells/well
  • CAL-148, MFM-223 and MDA-MB-453 cells were plated (10,000 cells/well) in quadruplicate into 96-well plates coated with 0.9% agarose diluted in corresponding media. Following 5 d of growth, cells were then treated with increasing half-log concentrations of GDC-0941 (10-3000 nM), GDC0980 (1 nM ⁇ 300 nM), BKM120 (10 nM-3000 nM) and NVP-BEZ235 (1 nM-300 nM) alone or in combination with 25 ⁇ M bicalutamide (CDX) for an additional 72 h, upon which cells were imaged and viability determined by Alamar blue.
  • FFPE Formalin fixed paraffin embedded tissue
  • AR Androgen receptor
  • p-AKT expression were evaluated in FFPE tissue using the DAKO (Carpinteria, CA) antibody: AR (clone AR411) at a 1:50 dilution and AKT p473 (Cell signaling, CA) at a 1:200 dilution for 1 h at room temperature.
  • FFPE tissue was subject to antigen retrieval with high pH buffer (pH 8.0) followed by overnight incubation with an AR (1:30) or Ki67 (1:75) antibody dilution overnight.
  • Cells were trypsinized, lysed and relative protein expression was determined by Western blot as previously described (JCI reference) with the following antibodies; the AR polyclonal antibody, SC-N20 (Santa Cruz Biotechnology), pS6-Ser235/236 (Cell Signaling #4856), S6 (Cell Signaling #2317), pAKT-5473 (Cell Signaling #9271), AKT (Cell Signaling #9272), PARP (Cell Signaling #9542) and GAPDH (Millipore).
  • the AR polyclonal antibody SC-N20 (Santa Cruz Biotechnology)
  • pS6-Ser235/236 Cell Signaling #4856
  • S6 Cell Signaling #231
  • pAKT-5473 Cell Signaling #9271
  • AKT Cell Signaling #9272
  • PARP Cell Signaling #9542
  • GAPDH Cell Signaling #9542
  • mice Five-week-old female athymic nude-Foxnlnu mice (Harlan Sprague-Dawley) were injected (s.c.) with either ⁇ 4 ⁇ 10 6 (CAL-148) or ⁇ 10 ⁇ 10 6 (MDA-MB-453) cells suspended in media (200 ⁇ L) into each flank using a 22-gauge needle. Once tumors reached a volume of 25-50 mm 3 , mice were randomly allocated to treatment or vehicle arms. Treatments included bicalutamide p.o. (100 mg/kg/d), GDC0980 p.o. (7.5 mg/kg/d) in 5% DMSO, 0.5% carboxymethyl cellulose (MCT) or NVP-BEZ235 p.o.
  • Tumor diameters were serially measured at the indicated times with digital calipers and tumor volumes were calculated as width 2 ⁇ length/2.
  • PDGFR inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines.
  • Bar graph displays the half-maximal inhibitory concentration (nM) of the PDGFR inhibitor sorafenib for a panel of TNBC cell lines.
  • IGF1R inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines.
  • Bar graph displays the half-maximal inhibitory concentration (nM) of the IGF1R inhibitor OSI-906 for a panel of TNBC cell lines.
  • IGF1R inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines.
  • Bar graph displays the half-maximal inhibitory concentration (nM) of the IGF1R inhibitor BMS-754807 for a panel of TNBC cell lines.
  • MET inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines.
  • Bar graph displays the half-maximal inhibitory concentration (nM) of the MET inhibitor PF2341066 for a panel of TNBC cell lines.
  • GSE-904 A USA Frozen tissue samples from primaiy, sporadic and 4 43 ER, PR, 17(40) 0.0 0.0 0.0 Richardson AL, et al. X chromosomal BRCA1-mutant breast obtained through the Harvard HER2 abnormalities in basal-like human breast Breast SPORE cancer. Cancer Cell . 2006; 9(2): 121-132.
  • GSE-2109 A,B USA The international genomics consortium collection of 351 Variable 0 60(17) breast cancer tissue samples processed by the expO biospecimen repository, obtained from community hospitals GSE-7390 A,D Europe Lymp node-negative breast cancer patients, systemically 198 ER 29(15) 2.5 Desmedt C, et al.
  • GSE-22513 A USA Invasive breast cancer pretreatrnent biopsies taken from 112 ER, PR, 25(22) 0.0 0.0 0.0 GSE-28821, patients at the Vanderbilt-Ingram Cancer Center HER2 GSE-28796 GSE-11121 A Germany Lymph node-negative breast cancer patients treated at 200 NA 19(10) Schmidt M, et al. The humoral immune system the Johannes Gutenberg University Mainz between 1988 has a key prognostic impact in node-negative and 1998. Patients were all treated with surgery without breast cancer. Cancer Res . 2008; 68(13): 5405- adjuvant therapy 5413.
  • GSE-2603 A USA Primary breast cancers surgically resected at the 99 ER, PR 32(32) 0.0 0.0 Minn AJ, et al. Genes that mediate breast Memorial Sloan-Kcttcring Cancer Center cancer metastasis to lung. Nature . 2005; 436(7050): 518-524. MDA133 F USA Tumors from prcopcrative paclitaxel and 5-FU, 133 ER, PR, 21(16) 0.0 3.0 0.8 Gibson GR, Qian D, Ku JK, Lai LL. doxonibicin. and CTX performed at the University HER2 Metaplastic breast cancer: clinical features and of Texas MD Anderson Cancer Center outcomes. Am Surg . 2005;71(9): 725-730.
  • GSE-5364 A Singapore Tumors from the National Cancer Centre of S 183 NA 31(17) Yu K. et al. A precisely regulated gene ingapore expression cassette potently modulates metastasis and survival in multiple solid cancers.
  • LAPTM4B adjuvant chemotherapy obtained from the HER2 and YWHAZ contributes to chemotherapy Harvard Breast SPORE, Boston, resistance and recurrence of breast cancer. Massachusetts, diagnosed between 2000 and Nat Med . 2010; 16(2): 214-218. 2003 GSE-20194 A USA
  • Pretreatment tumors obtained from stage I-III 278 ER, PR, 27(23) 2.6 4.3 0.0 Juul N, et al.
  • GSE-5327 and GSE-16446 data sets are derived from ER-negative tumors by IHC.
  • C GSE18864 data set includes only ER, PR and HER2 negative tuomrs by IHC.
  • B Molecular subtype determined by correlation with UNC/intrinsic breast centroids (29).
  • AC adenocarcinoma
  • ANC anaplastic carcinoma
  • ASCC acantholytic squamous cell carcinoma
  • C carcinoma
  • CS carcinosarcoma
  • DC ductal carcinoma
  • IDC invasive ductal carcinoma
  • IGA invasive galactopharic adenocarcinoma
  • INF inflammatory ductal carcinoma
  • MC metaplastic carcinoma
  • MBC medullaiy breast cancer.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Oncology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Biochemistry (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

In one aspect provided herein are methods of determining a triple negative breast cancer (TNBC) subtype in an individual in need thereof comprising determining expression of one or more genes in one or more TNBC cells of the individual; and comparing the expression of the one or more genes in the TNBC cells with the expression of the one or more genes in a control. In another aspect, the methods are directed to determining a treatment protocol for the TNBC patient based on the TNBC subtype. In another aspect, the methods are directed to predicting whether an individual will benefit from a treatment for a particular TNBC subtype. In yet another aspect, the invention is directed to a method of determining whether an agent can be used to treat a TNBC subtype.

Description

RELATED APPLICATION
This application is a continuation of U.S. application Ser. No. 14/358,330, filed May 15, 2014, which is a U.S. National Stage of International Application No. PCT/US2012/065724, filed Nov. 17, 2012, which claims the benefit of U.S. Provisional Application No. 61/561,743, filed on Nov. 18, 2011, the entire disclosures of which are incorporated herein by this reference.
GOVERNMENT SUPPORT
This invention was made with government support under grants CA95131, CA148375; CA105436 and CA070856, CA68485 and CA009385 awarded by the National Institute of Health (NIH). The government has certain rights in the invention.
SEQUENCE LISTING
The instant application contains a Sequence Listing which has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. The ASCII copy of the Sequence Listing, which was created on Jan. 17, 2020, is named 11672N-12047U.txt and is 2 kilobytes in size.
BACKGROUND OF THE INVENTION
Treatment of patients with triple-negative breast cancer (TNBC), lacking estrogen receptor (ER) and progesterone receptor (PR) expression as well as human epidermal growth factor receptor 2 (HER2) amplification, has been challenging due to the heterogeneity of the disease and the absence of well-defined molecular targets (Pegram M D, et al. J Clin Oncol. 1998; 16(8):2659-2671; Wiggans R G, et al. Cancer Chemother Pharmacol. 1979; 3(1):45-48; Carey L A, et al. Clin Cancer Res. 2007; 13(8):2329-2334). TNBCs constitute 10%-20% of all breast cancers, more frequently affect younger patients, and are more prevalent in African-American women (Morris G J, et al. Cancer. 2007; 110(4):876-884). TNBC tumors are generally larger in size, are of higher grade, have lymph node involvement at diagnosis, and are biologically more aggressive (Haffty B G, et al. J Clin Oncol. 2006; 24(36):5652-5657). Despite having higher rates of clinical response to presurgical (neoadjuvant) chemotherapy, TNBC patients have a higher rate of distant recurrence and a poorer prognosis than women with other breast cancer subtypes (Haffty B G, et al. J Clin Oncol. 2006; 24(36):5652-5657; Dent R, et al. Clin Cancer Res. 2007; 13(15 pt 1):4429-4434). Less than 30% of women with metastatic TNBC survive 5 years, and almost all die of their disease despite adjuvant chemotherapy, which is the mainstay of treatment (Dent R, et al. Clin Cancer Res. 2007; 13(15 pt 1):4429-4434).
One of the first molecular insights into TNBCs was the observation that they are likely to arise in BRCA1 mutation carriers and have gene expression (GE) profiles similar to those of BRCA1-deficient tumors (Haffty B G, et al. J Clin Oncol. 2006; 24(36):5652-5657). BRCA1 plays an important role in DNA double-strand break repair, contributing to the maintenance of DNA stability (D'Andrea A D, Grompe M. Nat Rev Cancer. 2003; 3(1):23-34). Poly ADP-ribose polymerase (PARP) enzymes are critical for appropriate processing and repair of DNA breaks (Hoeijmakers J H. Nature. 2001; 411(6835):366-374). Tumor cell lines lacking functional BRCA1 or BRCA2 are sensitive to PARP inhibitors in preclinical studies (Farmer H, et al. Nature. 2005; 434(7035):917-921). Clinical trials using both PARP inhibitors and DNA-damaging agents (e.g., cisplatin) in TNBC are currently underway and show promise in BRCA1/2-mutant tumors (Fong P C, et al. N Engl J Med. 2009; 361(2):123-134). Other studies identifying molecular markers of TNBC, such as VEGF (Burstein H J, et al. J Clin Oncol. 2008; 26(11):1810-1816), EGFR (Nielsen T O, et al. Clin Cancer Res. 2004; 10(16):5367-5374), Src (Finn R S, et al. Breast Cancer Res Treat. 2007; 105(3):319-326), and mTOR (Ellard S L, et al. J Clin Oncol. 2009; 27(27):4536-4541) have been important for the design of clinical trials investigating targeted treatments.
Clearly, there is a major need to better understand the molecular basis of TNBC and to develop effective treatments for this aggressive type of breast cancer.
SUMMARY OF THE INVENTION
In one aspect provided herein are methods of determining a triple negative breast cancer (TNBC) subtype in an individual in need thereof comprising determining expression of one or more genes (e.g., presence of one or more mRNAs and/or protein encoded by gene) in one or more TNBC cells of the individual; and comparing the expression of the one or more genes in the TNBC cells with the expression of the one or more genes in a control. Increased expression of one or more genes comprising one or more cell cycle genes, cell division genes, proliferation genes, DNA damage response genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC BL1 subtype. Increased expression of one or more growth factor signaling genes, glycolysis genes, gluconeogenesis genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC BL2 subtype. Increased expression of one or more immune cell signaling genes, cytokine signaling genes, antigen processing and presentation genes, core immune signal transduction genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC IM subtype. Increased expression of one or more cell motility genes, extracellular matrix (ECM) receptor interaction genes, cell differentiation genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC M subtype. Increased expression of one or more cell motility genes, cellular differentiation genes, growth pathway genes, growth factor signaling genes, angiogenesis genes, immune signaling genes, stem cell genes, HOX genes, mesenchymal stem cell-specific marker genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC MSL subtype. Increased expression of one or more hormone regulated genes in the TNBC cells compared to the control indicates that the TNBC is a TNBC LAR type.
In another aspect, the methods are directed to methods of determining a treatment protocol for the triple negative breast cancer (TNBC) patient based on the TNBC subtype.
In another aspect, the methods are directed to predicting whether an individual (e.g., patient) will benefit from a (one or more) treatment for a particular TNBC subtype.
In yet another aspect, the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) subtype comprising contacting a cell line that is a model for the TNBC subtype of interest with an agent to be assessed; and determining viability of the cell line in the presence of the agent, wherein if the viability of the cell line decreases in the presence of the agent, then the agent can be used to treat the TNBC subtype of interest. The TNBC subtype of interest can be a TNBC BL-1 subtype, a TNBC BL-2 subtype, a TNBC IM subtype, a TNBC M subtype, a TNBC MSL subtype or a TNBC LAR subtype.
BRIEF DESCRIPTION OF THE DRAWINGS
FIGS. 1A-1C: Filtering GE data sets to identify TNBCs. (1A) Flow chart of analysis. Human breast cancer GE profiles (training=2353, validation=894) were normalized within individual data sets and a bimodal filter was applied to select ER, PR, and HER2 negative samples by GE, resulting in 386 samples in the training set and 201 samples in the validation set with a triple-negative phenotype. k-means clustering was performed on the training set, and a GE signature representing the TNBC subtypes from the training set was used to predict the best-fit subtype for each TNBC profile in an independent validation set. GSE-A was performed on the training and validation sets to identify enriched canonical pathways for each TNBC subtype. (1B) Histograms show the distribution and frequency of tumors using relative ER, PR, and HER2 GE levels (log 2) and bimodal fit to identify TN tumor samples. Dashed line indicates the expression value at the center of the positive expression peak used to select controls for C. (1C) Heat map representation of GE for 386 TNBCs relative to 5 IHC-validated controls for each ER, PR, and HER2.
FIGS. 2A-2E: Identification of TNBC subtypes. (2A) Silhouette plot showing the composition (n=number of tumors) and stability (AVG width) of k-means clustering on the TNBC training set. Clusters with s(I)>0 were considered stable. (2B) Consensus clustering displaying the robustness of sample classification using multiple iterations (×1000) of k-means clustering. (2C) The CDF depicting the cumulative distribution from consensus matrices at a given cluster number (k). (2D) The optimal cluster number is 7 at the point in which the relative change in area (Δ) under the CDF plot does not change with increasing k. (2E) Principal component analysis graphically depicting fundamental differences in GE between the TNBC clusters. The cluster (subtypes) are named as shown.
FIG. 3 : GE patterns within TNBC subtypes are reproducible. Heat maps showing the relative GE (log 2, −3 to 3) of the top differentially expressed genes (P<0.05) in each subtype in the training set (left) and the same differentially expressed genes used to predict the best-fit TNBC subtype of the validation set (right). Overlapping gene ontology (GO) terms for top canonical pathways in both the training and validation sets as determined by GSE-A are shown to the right of the heat maps.
FIGS. 4A-4F: Basal-like TNBC subtypes have differential sensitivity to DNA-damaging agents. IC50 values for TNBC cell lines treated with PARP inhibitors (4A) veliparib, (4C) olaparib, or (4E) cisplatin for 72 hours. Error bars reflect SEM for 3 independent experiments. Black horizontal lines above various bars in the plots indicate cell lines that failed to achieve an IC50 at the highest dose of veliparib (30 μM), olaparib (100 μM), or cisplatin (30 μM). Cell lines that carry BRCA1 or BRCA2 mutations (pink) are displayed below the graph. Dot plot shows the log distribution of drug sensitivity to PARP inhibitors (B) veliparib, (4D) olaparib, or (4F) cisplatin in the basal-like subtypes (BL=BL1+BL2), the mesenchymal-like subtypes (ML=M+MSL), and the LAR subtype. Black horizontal bars in the dot plot indicate the mean IC50 for each of the subtypes. *Statistically significant differences in IC50 values of BL compared with ML (P=0.017) and LAR (P=0.032), as determined by Mann-Whitney U test.
FIGS. 5A-5D: Differential sensitivity of the LAR TNBC subtype to AR and Hsp90 inhibitors. IC50 values for each TNBC cell line after treatment with (5A) bicalutamide or (5C) the Hsp90 inhibitor 17-DMAG for 72 hours. Black bar above bicalutamide indicates cell lines that failed to achieve an IC50. Heat map displays relative AR expression (log 2) across TNBC cell lines. Dot plot shows log distribution of drug sensitivity to (5B) bicalutamide or (5D) 17-DMAG in the basal-like (BL=BL1+BL2), mesenchymal-like (ML=M+MSL), and LAR subtypes. Black horizontal bars in the dot plot indicate the mean IC50 for each of the subtypes. *Statistically significant differences in IC50 values of LAR versus BL (P=0.007) or ML (P=0.038) after bicalutamide and LAR versus BL and ML (P=0.05) after 17-DMAG treatments, as determined by Mann-Whitney U test.
FIGS. 6A-6D: Mesenchymal-like TNBC subtypes are sensitive to dasatinib and NVP-BEZ235. IC50 values for each TNBC cell lined treated with (6A) dasatinib or (6C) NVP-BEZ235 for 72 hours. Cell lines that have PIK3CA mutations (red) or are deficient in PTEN (blue, circle indicates mutated) are displayed below the NVP-BEZ235 graph. Dot plots show the log distribution of drug sensitivity to (6B) dasatinib or (6D) NVP-BEZ235 in the basal-like subtypes (BL=BL1+BL2), mesenchymal-like subtypes (ML=M+MSL), and LAR. Black horizontal bars in the dot plots indicate the mean IC50 for each of the subtypes. *Statistically significant differences in IC50 values of BL versus ML (P=0.020) when treated with dasatinib and ML versus BL (P=0.001) and LAR versus BL (P=0.01) when treated with NVP-BEZ235, as determined by Mann-Whitney U test.
FIG. 7 : Xenograft tumors established from TNBC subtypes display differential sensitivity to cisplatin, bicalutamide, and NVP-BEZ235. Nude mice bearing established tumors (25-50 mm3) from basal-like (HCC1806 and MDA-MB-468), LAR (SUM185PE and CAL-148), or mesenchymallike (CAL-51 and SUM159PT) were treated with cisplatin (red), bicalutamide (purple), NVP-BEZ235 (green), or vehicle (blue) for approximately 3 weeks. Serial tumor volumes (mm3) were measured at the indicated days. Each data point represents the mean tumor volume of 16 tumors; error bars represent SEM.
FIG. 8 : Random distribution of TNBC subtypes found within datasels. The percent of tumors in each cluster is displayed across 14 studies that comprise the training dataset.
FIG. 9 : Consensus clustering of training and validation TNBC datasets (n=587). Heat map displays consensus clustering results depicting the robustness of sample classification. Red areas indicate samples that frequently cluster with each other over multiple iterations (1000) of k-means clustering (k::c 2 to k=9).
FIG. 10 : Gene expression profiles derived from tumors that were laser-capture microdissected represent all TNBC subtypes. Bar graph depicts the percentage of tumors that were microdissected (blue bars) present in the GSE584 (n=17) and Vanderbilt (n=14) datasets relative to non-microdissected tumors in the combined dataset (red bars).
FIGS. 11A-11B: TNBC subtypes identified by IHC. (11A) Silhouette plot showing the composition (n=number of tumors) and stability (AVG width) of k-means clustering on the TNBC training sel. Clusters with a silhouette width, s(i)>O were considered stable. (11B) Heatmap displays hierarchical clustering of ERBB2, PGR, ESR1 and AR expression in the tumors identified by IHC. Color bar identifies the cluster associated with each tumor.
FIG. 12 : TNBC subtypes Identified by IHC display similar GE patterns to the six TNBC subtypes. IHC TNBC clusters are shown in silhouette plot with relative GE for genes unique to the six TNBC subtypes shown below.
FIG. 13 : Differential GE across TNBC subtypes. Heatmaps show relative GE (log 2, −2 to 2) associated with proliferation, DNA damage response, myoepithelial genes, immune signal transduction, TGFβ signaling, growth factor receptors, EMT, WNT signaling, stem-like, claudin (CL), angiogenesis, AR and of AR target genes across TNBC subtypes (as in FIG. 3 ).
FIGS. 14A-14B: TNBC tumor subtypes differentially stain for the proliferation marker Ki-67. (14A) Representative micrographs from 20 tumors showing IHC staining of the proliferation marker, Ki-67, in tumors from different TNBC subtypes. (14B) Dol plot showing the mean and distribution of Ki-67 staining within TNBC subtypes as scored by study pathologist. Subtypes: BL1=basal-like 1, BL2=basal-like 2, IM=immunomodulatory, M=mesenchymal-like, MSL=mesenchymal stem-like and LAR=luminal AR.
FIG. 15 : Response rates differ across TNBC subtypes in taxane-treated patients. Percent of patients achieving pathologic complete response (pCR) after taxane-based treatment was significantly (P=0.042, chi-squared analysis) different for patients whose tumors correlated to basal-like (n=19), mesenchymal-like (n=16) and luminal AR (n=7) subtypes, from studies in which response data were available.
FIG. 16 : 1M subtype Is enriched in Immune cell signaling. Heatmaps showing the relative GE (log 2, −2 to 2) for genes involved in immune cell surface antigens, cytokines, immune cell signal transduction, complement, chemokine, and antigen presentation across TNBC subtypes (as in FIG. 3 ).
FIG. 17 : The claudin-Iow predictor gene set identifies a sub-population of MSL tumors. Unsupervised hierarchical clustering was performed on the training TNBC tumors using genes (n=770) unique to the claud in•low subgroup [26]. Displayed to the right of the heatmap are the genes that are most differentially expressed (either high or low) in the claudin-Iow tumor set. Colorbar displays the TNBC subtype and heatmaps show relative levels (Log 2) 01 claudins (3, 4 and 7) and CD24, markers of this subgroup.
FIGS. 18A-18B: TNBC tumor subtypes differentially stain for AR by IHC. (18A) IHC staining for AR from 20 tumors with representative samples from each TNBC subtype shown. (18B) Dot plot showing the quantification of nuclear AR staining based on intensity and the percent of nuclei staining positive for AA in 20 tumors; Note, in some cases one dot represents overlapping dots from multiple tumors.
FIGS. 19A-19B: An androgen-inducible gene signature segregates LAR tumors. Hierarchical clustering was performed on both the (19A) training and (19) validation TNBC tumor sel using a 559 androgen-inducible gene signature (Hayes M J et al. Clin Cancer Res. 2008; 14(13):4038-4044).
FIGS. 20A-20B: TNBC subtypes differentially correlate with the intrinsic molecular subtypes. (20A) Heatmaps show relative GE (log 2, −2 to 2) of luminal and basal markers of breast cancer across all TNBC subtypes. (20B) Bar graph shows the distribution of intrinsic molecular subtypes of breast cancer (luminal A and B, normal breast-like, HER2, basal-like or unclassified) within each TNBC subtype (UNS=unstable, BL1=basal-like 1, B12=basal-like 2, 1M=immunomodulatory, M=mesenchymal-like, MSL=mesenchymal stem-like and LAR=luminal AR) as determined by best-fit Spearman correlation to the intrinsic centroids.
FIGS. 21A-21D TNBC subtypes differ in relapse-free survival and distant metastasis-free survival. Kaplan-Meier plot showing (21A) to-year RFS or (21C) DMFS in TNBC subtypes. RFS (21B) and DMFS (21D) hazard ratios (bold) and 95% CI (parentheses) for patients from TNBC subtypes. Shaded boxes indicate significant (P<0.05) comparisons.
FIGS. 22A-22B: TNBC subtypes differ in age, but are similar size upon diagnosis. Box plots show the median (horizontal line), range (rectangle) and SD (error bars) of (22A) age at diagnosis and (22B) tumor size (mm) between TNBC subtypes (as in FIG. 3 ); •P=9.0e-6
FIGS. 23A-23B: Chromosomal Aberrations in TNBC cell lines. (23A) Box plots depicting the average number of chromosomes (mode) in breast cancer cell lines correlating to basal-like (n=8) vs. mesenchymal-like (n=6) subtypes. (23B) Box plots depicting the average number of chromosomal rearrangements (translocations, inversions, and deletions) in basal-like vs. mesenchymal-like subtypes. Chromosome mode and rearrangements were obtained from Departments of Pathology and Oncology, Univ of Cambridge (www.path.cam.ac.uk/˜pawefish/cell%20line%20catalogues/breast-cell-lines.htm). *P<0.01 by unpaired t-test.
FIG. 24 : TNBC cell lines cluster in three major groups: luminal AR, basal-like and mesenchymal-like. Unsupervised hierarchical clustering of TNBC cell lines performed on genes unique to TNBC subtypes (n=2188). Colorbar shows the best correlation of cell lines to the TNBC subtypes.
FIGS. 25A-25C: LAR cell lines depend on AR expression for colony formation. (25A) Top panel depicts relative AR mRNA levels obtained from GE microarrays performed on TNBC cell lines (log 2, centered on 0). Color bar identifies TNBC subtype classification for each cell line (immunomodulatory shown in orange and unclassified shown in red). Immunoblots showing relative expression AR protein in TNBC cell lines, Hsp90 serves as a loading control. (25B) AR expression 72 h following transfection with siRNA pools of non-targeting (NT) or targeting AR in MFM-223, MDA-MB-453 and SUM185PE cells. GAPDH expression serves as a loading control. (25C) Colony formation of MFM-223, MDA-MB-453 and SUM185PE cells 14d following siRNA transfection of AR or non-targeting control (NT).
FIGS. 26A-26B: Identification of PIK3CA mutations in AR-positive TNBC cell lines and tumors. (26A) Detection of PIK3CA 1047 mutation using digital droplet PCR. DNA from cell lines with varying PIK3CA status (HMEC=H1047R wt/wt, MDAMB453=H1047R wt/mut, SUM185=H1047R mut/mut and SUM159=H1047L wt/mut) were simultaneously amplified with fluorescent-conjugated primers to wild-type (VIC) or mutant (FAM) and the percentage of wt vs. mut DNA was measured by digital droplet PCR. DNA that did not undergo PCR amplification is indicated by no reaction (NR). (26B) Heatmap displays the TNBC molecular subtype based of TCGA samples with corresponding levels of AR RNA (RNA-seq) and protein (RPPA) for AR and phosphorylated AKT (T308 and S473). Color bar indicates PIK3CA mutations (red) within TNBC from the TCGA.
FIG. 27 : AR-expressing TNBC cells display active PI3K pathway. Heatmap displays relative protein levels (RPPA) of AR, AKT (S473 and T308), GSK3β (S9 and S21) and PTEN across a large panel of TNBC cell lines. Hierarchical clustering was performed on cell lines and revel clusters of cell lines with active PI3K through PTEN loss or PIK3CA mutations.
FIGS. 28A-28C-2 : AR-positive TNBC cell lines display active PI3K pathway and are sensitive to PI3K inhibitors. (28A) Immunoblot displays relative levels of AR, p-AKT, p-S6 in AR-dependent prostate cancer (LNCaP), human mammary epithelial cells (HMEC) and LAR TNBC, with GAPDH serving as a loading control. (28B) IHC-stained sections display cellular levels of AR, and p-AKT in cell lines from the same tissue microarray (TMA). (28C-1 and 28C-2) Bar graphs display the 50% inhibitory concentration (IC50) for TNBC cell lines treated with single-agent pan PI3K inhibitors (BKM-120 and GDC-0941) (28C-1) or PI3K/mTOR inhibitors (NVP-BEZ235 or GDC-0980) (28C-2) for 72 h. Black bars above graphs indicate cell lines in which the IC50 was not reached at maximal dose. Bar below indicates normal HMEC cells and cell lines carrying PIK3CA mutations.
FIGS. 29A-29B: Genetic targeting of AR increases the efficacy of the PI3K inhibitor GDC-0941 and PI3K/mTOR inhibitor GDC-0980. (29A) Immunoblot displays decreased AR expression at 72 h following infection with two shRNAs targeting AR (shAR1 and shAR2) compared to infection with nontargeting shRNA (shNT) in three AR-positive TNBC cell lines. Actin levels below serve as loading controls. (29B) Line graphs display relative viability of LAR cell lines infected with nontargeting (shNT) or shRNAs targeting AR (shAR1 and shAR2) following a 72 h treatment with the pan-PI3K inhibitor GDC-0941 (top) or the dual PI3K/mTOR inhibitor GDC-0980.
FIGS. 30A-30B: Pharmacological targeting of AR with bicalutamide increases the efficacy of GDC0941 and GDC0980 in AR-expressing TNBC. (30A) Line graphs show relative viability of AR-expressing cell lines treated with increasing doses of GDC-0941 (top) or GDC-0980 (bottom) alone or in combination with 25 μM bicalutamide (CDX). Dashed black line depicts the theoretical line of additivity of both drugs determined from the effect of bicalutamide alone and either GDC-0941 or GDC-0980 alone. (30B) Immunoblots from three AR-expressing TNBC cell lines treated with either bicalutamide (CDX), GDC-0941, GDC0980 alone or bicalutamide in combination with either GDC-0941 or GDC-0980 for 24 h or 48 h were probed for AR, PARP, p-AKT, AKT, p-S6, S6 and GAPDH.
FIGS. 31A-31B: Combined inhibition of AR and PI3K increases apoptotic cell death in AR+TNBC cell lines. (31A) Bar graphs display relative caspase 3/7 activity (RLU) normalized to viable cell number 48 hrs after treatment with vehicle, positive control (3 μM adriamycin, ADR), 50 μM bicalutamide (CDX) and 3 μM GDC-0941 or 1 μM GDC-0980 alone or in combination with CDX. (31B) Histograms show representative cell cycle analysis of MDA-MB-453 cells treated with similar conditions as above. Indicated percentages represent the fraction of hypodiploid DNA (sub-G1), indicative of late stage apoptotic DNA fragmentation.
FIGS. 32A-32C: Simultaneous targeting of AR and PI3K decreases viability of AR-expressing cell lines grown in a 3-D forced suspension assay and inhibits in vivo xenograft growth. (31A) Representative images display 3-D cell aggregates of MDA-MB-453 cells treated with increasing doses of GDC-0941 or GDC-0980 in the absence or presence of 25 μM bicalutamide (CDX). (32B) Line graphs display relative viability of 3-D cell aggregates treated with GDC-0941 or GDC-0980 alone or in combination with 25 μM bicalutamide (CDX). Dashed black line depicts the theoretical line of additivity determined from the effect of bicalutamide alone and either GDC-0941 or GDC-0980 alone. (32C) Nude mice bearing established tumors from AR-positive TNBC cell lines (CAL-148 and MDA-MB-453) were treated with either bicalutamide (CDX, black hashed line), NVP-BEZ235, GDC-0980 or with the combination of bicalutamide and either NVP-BEZ235 (blue hashed line) or GDC-0980 (red hashed line). Serial tumor volumes (mm3) were measured at the indicated days. Each data point represents mean tumor volume of 16 tumors; error bars represent SEM.
FIGS. 33A-33B: Detection of PIK3CA mutations by Sanger and digital droplet PCR. (33A) Electropherograms display representative results from sanger sequencing of the E20 amplicon in a PIK3CA wild-type (top) or a H1047R PIK3CA mutant sample. (33B) Scatterplot shows the percent of cells with mutant PIK3CA detected by digital droplet compared to Sanger sequencing.
FIG. 34 : Genetic targeting of AR increases the efficacy of the PI3K inhibitor BKM-120 and PI3K/mTOR inhibitor NVP-BEZ235. Line graphs display relative viability of LAR cell lines infected with nontargeting (shNT) or shRNAs targeting AR (shAR1 and shAR2) following a 72 h treatment with the pan-PI3K inhibitor BKM-120 (top) or the dual PI3K/mTOR inhibitor NVP-BEZ235.
FIG. 35 : Pharmacological targeting of AR with bicalutamide increases the efficacy of BKM120 and NVP-BEZ235 in AR-expressing TNBC. Line graphs show relative viability of AR-expressing cell lines treated with increasing doses of BKM120 (top) or NVP-BEZ235 (bottom) alone or in combination with 25 μM bicalutamide (CDX). Dashed black line depicts the theoretical line of additivity determined from the effect of bicalutamide alone and either BKM120 or NVP-BEZ235 alone.
FIGS. 36A-36B: Simultaneous targeting of AR and PI3K decreases viability of AR-expressing cell lines grown in a 3-D forced suspension assay. (36A) Line graphs display relative viability of 3-D cell aggregates treated with BKM120 or NVP-BEZ235 alone or in combination with 25 uM bicalutamide (CDX). Dashed black line depicts the theoretical line of additivity determined from the effect of bicalutamide alone and either BKM120 or NVP-BEZ235 alone. (36B) Immunoblots from three AR-expressing TNBC cell lines treated with either bicalutamide (CDX), BKM-120, BEZ235 alone or in combination with bicalutamide for 24 h or 48 h. Western blots were probed for AR, PARP, p-AKT, AKT, p-S6, S6 and GAPDH.
FIG. 37 : Combined Targeting of PI3K and AR increase apoptosis detected by annexin V and propidium iodide (PI) staining. Scattergrams show the percentage of viable (lower left quadrant, annexin V and PI negative), early apoptotic (lower right quadrant, annexin V-positive and PI-negative), or late apoptotic/necrotic (upper right quadrant, annevin V and PI positive) cells following 48 hrs of treatment with either GDC-0941 or GDC-0980 alone or in combination with 50 μM bicalutamide (CDX).
FIGS. 38A-38C: Genetic and pharmacological targeting of AR increases p-AKT.
FIG. 39 : PDGFR inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines. Bar graph displays the half-maximal inhibitory concentration (nM) of the PDGFR inhibitor sorafenib for a panel of TNBC cell lines. Colorbar below designates the TNBC subtype (basal-like=black, mesenchymal-like=grey) or normal cells (white).
FIG. 40 : IGF1R inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines. Bar graph displays the half-maximal inhibitory concentration (nM) of the IGF1R inhibitor OSI-906 for a panel of TNBC cell lines. Colorbar below designates the TNBC subtype (basal-like=black, mesenchymal-like=grey) or normal cells (white).
FIG. 41 : IGF1R inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines. Bar graph displays the half-maximal inhibitory concentration (nM) of the IGF1R inhibitor BMS-754807 for a panel of TNBC cell lines. Colorbar below designates the TNBC subtype (basal-like=black, mesenchymal-like=grey) or normal cells (white).
FIG. 42 : MET inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines. Bar graph displays the half-maximal inhibitory concentration (nM) of the MET inhibitor PF2341066 for a panel of TNBC cell lines. Colorbar below designates the TNBC subtype (basal-like=black, mesenchymal-like=grey) or normal cells (white).
DETAILED DESCRIPTION OF THE INVENTION
Triple-negative breast cancer (TNBC) is a highly diverse group of cancers, and subtyping is necessary to better identify molecular-based therapies. In this study, gene expression (GE) profiles from 21 breast cancer data sets were analyzed and 587 TNBC cases were identified. Cluster analysis identified 6 TNBC subtypes displaying unique GE and ontologies, including 2 basal-like (BL1 and BL2), an immunomodulatory (IM), a mesenchymal (M), a mesenchymal stem-like (MSL), and a luminal androgen receptor (LAR) subtype. Further, GE analysis allowed identification of TNBC cell line models representative of these subtypes. Predicted “driver” signaling pathways were pharmacologically targeted in these cell line models as proof of concept that analysis of distinct GE signatures can inform therapy selection. BL1 and BL2 subtypes had higher expression of cell cycle and DNA damage response genes, and representative cell lines preferentially responded to cisplatin. M and MSL subtypes were enriched in GE for epithelial-mesenchymal transition, and growth factor pathways and cell models responded to NVP-BEZ235 (a PI3K/mTOR inhibitor) and dasatinib (an abl/src inhibitor). The LAR subtype includes patients with decreased relapse-free survival and was characterized by androgen receptor (AR) signaling. LAR cell lines were uniquely sensitive to bicalutamide (an AR antagonist). These data provide biomarker selection, drug discovery, and clinical trial design that align TNBC patients to appropriate targeted therapies.
An extensive number of TNBC GE profiles was compiled with the intent of molecularly subtyping the disease. Six (6) TNBC subtypes were identified. Further, using GE signatures derived from each TNBC subtype, representative TNBC cell lines that serve as models for the different subtypes of the disease were aligned. Using the panel of cell lines, prominent signaling pathways revealed by GE signatures were pharmacologically targeted and it was found that the cell lines representing the various subtypes had different sensitivities to targeted therapies currently under laboratory and clinical investigation. The identification of diverse TNBC subtypes and the molecular drivers in corresponding cell line models provides great insight to the heterogeneity of this disease and provides preclinical platforms for the development of effective treatment (see Lehmann, B. D., et al., J Clin Invest, 121(7): 2750-2767 (July 2011), the entire teachings of which are incorporated herein by reference).
Also described herein is further investigation of a panel of AR-positive TNBC tumors which showed that PIK3CA kinase mutations are a frequent event in the LAR subtype (65.3% vs. 38.4%) and were not just selected for in vitro during establishment of the tumor-derived cell lines. Also shown herein is that PI3K pathway activation plays a role in resistance to AR antagonists, as there are elevated levels of activated AKT in residual tumors after bicalutamide treatment of xenograft tumors. Further, it was found that genetic or pharmacological targeting of AR synergizes with PI3K/mTOR inhibition in both two- and three-dimensional cell culture models. Based on the cell culture-based results, the combination of these inhibitors in in vivo xenograft studies was examined using bicalutamide +/−GDC0980 or NVP-BEZ235. The preclinical data herein provide the rationale for pre-selecting AR-positive TNBC patients for treatment with the combination of AR antagonists and/or PI3K/mTOR inhibitors.
As discussed herein, triple negative breast cancer (TNBC) refers to breast cancers that are estrogen receptor (ER) negative, progesterone receptor (PR) negative and human epidermal growth factor receptor 2 (HER-2) negative. The invention is based, in part, of the discovery of six TNBC subtypes.
Accordingly, in one aspect provided herein are methods of determining a triple negative breast cancer (TNBC) subtype in an individual in need thereof comprising determining expression of one or more genes (e.g., presence of one or more mRNAs and/or protein encoded by gene) in one or more TNBC cells of the individual; and comparing the expression of the one or more genes in the TNBC cells with the expression of the one or more genes in a control. Increased expression of one or more genes comprising one or more cell cycle genes, cell division genes, proliferation genes, DNA damage response genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC BL1 subtype. Increased expression of one or more growth factor signaling genes, glycolysis genes, gluconeogenesis genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC BL2 subtype. Increased expression of one or more immune cell signaling genes, cytokine signaling genes, antigen processing and presentation genes, core immune signal transduction genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC IM subtype. Increased expression of one or more cell motility genes, extracellular matrix (ECM) receptor interaction genes, cell differentiation genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC M subtype. Increased expression of one or more cell motility genes, cellular differentiation genes, growth pathway genes, growth factor signaling genes, angiogenesis genes, immune signaling genes, stem cell genes, HOX genes, mesenchymal stem cell-specific marker genes or a combination thereof in the TNBC cells compared to the control indicates that the TNBC is a TNBC MSL subtype. Increased expression of one or more hormone regulated genes in the TNBC cells compared to the control indicates that the TNBC is a TNBC LAR type.
In a particular aspect, the invention is directed to a method of determining a TNBC BL1 subtype in an individual in need thereof comprising determining increased expression of one or more cell cycle genes, cell division genes, proliferation genes, DNA damage response genes or a combination thereof in the TNBC cells of the individual compared to the control. Increased expression of the one or more genes indicates that the TNBC is a BL1 subtype TNBC. In one aspect, the method of determining a TNBC BL1 subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising AURKA, AURKB, CENPA, CENPF, BUB1, BUB1B, TTK, CCNA2, PLK1, PRC1, MYC, NRAS, PLK1, BIRC5, CHEK1, FANCA, FANCG, FOXM1, HNGA1, RAD54BP, RAD51, NEKS, NBN, EXO1, MSH2, MCM10, RAD21,SIX3, Z1C1, SOX4, SOX1 and MDC1 wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC BL1 subtype. Determination that the TNBC is a TNBC BL1 subtype can further comprise detecting increased expression of Ki-67 mRNA in the TNBC cells compared to the control. In addition, determination that the TNBC is a TNBC BL1 subtype can further comprise detecting one or more mutated genes. Examples of such mutated genes include mutated BRCA1, STAT4, UTX, BRCA2, TP53, CTNND1, TOP2B, CAMK1G, MAPK13, MDC1, PTEN, RB1, SMAD4, CDKN2A, ATM, ATR, CLSPN, HDAC4, NOTCH1, SMARCAL1, and TIMELESS.
In another aspect, the invention is directed to a method of determining a TNBC BL2 subtype in an individual in need thereof comprising determining increased expression of one or more growth factor signaling genes, glycolysis genes, gluconeogenesis genes or a combination thereof in the TNBC cells compared to the control. Increased expression of the one or more genes indicates that the TNBC is a TNBC BL2 subtype. In one aspect, the method of determining a TNBC BL subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising EGFR, MET, ELF4, MAF, NUAK1, JAG1, FOSL2, 1D1, ZIC1, SOX11, 1D3, FHL2, EPHA2, TP63 and MME wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC BL2 subtype. Determination that the TNBC is a TNBC BL2 subtype can further comprise detecting one or more mutated genes. Examples of such genes comprise mutated BRCA1, RB1, TP53, PTEN, CDKN2A, UTX, BRAC2, PTCH1, PTCH2, and RET.
In another aspect, the invention is directed to a method of determining a TNBC M subtype in an individual in need thereof comprising determining increased expression of one or more cell motility genes, extracellular matrix (ECM) receptor interaction genes, cell differentiation genes or a combination thereof in the TNBC cells compared to the control. Increased expression of the one or more genes indicates that the TNBC is a TNBC M subtype. In one aspect, the method of determining a TNBC M subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising TGFB1L1, BGN, SMAD6, SMAD7, NOTCH1, TGFB1, TGFB2, TGFB3, TGFBR1, TGFBR2, TGFBR3, MMP2, ACTA2, SNAI2, SPARC, SMAD7, PDGFRA, TAGLN, TCF4, TWIST1, ZEB1, COL3A1, JAG1, EN1, MYLK, STK38L, CDH11, ETV5, IGF1R, FGFR1, FGFR2, FGFR3, TBX3, COL5A2, GNG11, ZEB2, CTNNB1, DKK2, DKK3, SFPR4, TCF4, TCF7L2, FZD4, CAV1, CAV2, CCND1 and CCND2 wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC M subtype. Determination that the TNBC is a TNBC M subtype can further comprise detecting decreased E-cadherin (CDH1) expression compared to the control. Determination that the TNBC is a TNBC M subtype can further comprise detecting one or more mutated genes. Examples of such mutated genes include mutated PTEN, RB1, TP53, PIK3CA, APC, BRAF, CTNNB1, FGFR1, GLI1, HRAS, KRAS, NOTCH1, and NOTCH4. Determination that the TNBC is a TNBC M subtype can further comprise detecting chromosomal amplifications in KRAS, IGF1R or MET.
In another aspect, the invention is directed to a method of determining a TNBC MSL subtype in an individual in need thereof comprising determining increased expression of one or more cell motility genes, cellular differentiation genes, growth pathway genes, growth factor signaling genes, angiogenesis genes, immune signaling genes, stem cell genes, HOX genes, mesenchymal stem cell-specific marker genes or a combination thereof in the TNBC cells compared to the control. Increased expression of the one or more genes indicates that the TNBC is a TNBC MSL subtype. In one aspect, the method of determining a TNBC MSL subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising VEGFR2 (KDR), TEK, TIE1, EPAS1, ABCA8, PROCR, ENG, ALDHA1, PER1, ABCB1, TERF2IP, BCL2, BMP2, EPAS1, STAT4, PPARG, JAK1, ID1, SMAD3, TWIST1, THY1, HOXA5, HOXA10, GL13, HHEX, ZFHX4, HMBOX1, FOS, PIK3R1, MAF, MAFB, RPS6KA2, TCF4, TGFB1L1, MEIS1, MEIS2, MEOX1, MEOX2, MSX1, ITGAV, KDR, NGFR, NTSE, PDGFRA, PDGFRB, POU2F1, and VCAM1 wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC MSL subtype. Determination that the TNBC is a TNBC MSL subtype can further comprise detecting low expression of claudins 3, 4, 7 or a combination thereof compared to the control. Determination that the TNBC is a MSL subtype TNBC can further comprise detecting one or more mutated genes. Examples of such mutated genes include mutated CDKN2A, HRAS, TP53, NF1, PIK3CA, BRCA1, BRAF, KRAS, NF2, PDGFRA, APC, CTNNB1, FGFR1, PDGFRB.
In another aspect, the invention is directed to a method of determining a TNBC IM subtype in an individual in need thereof comprising determining increased expression of one or more immune cell signaling genes, cytokine signaling genes, antigen processing and presentation genes, core immune signal transduction genes or a combination thereof in the TNBC cells compared to the control. Increased expression of the one or more genes indicates that the TNBC is a TNBC IM subtype. In one aspect, the method of determining a TNBC IM subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising CCL19, CCL3, CCL4, CCL5, CCL8, CCR1, CCR2, CCR5, CCR7, CD2, CD37, CD38, CD3D, CD48, CD52, CD69, CD74, and CD8A wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC IM subtype. Determination that the TNBC is a TNBC IM subtype can further comprise detecting one or more mutated genes. Examples of such mutated genes include TP53, CTNNA1, DDX18, HUWE1, NFKBIA, APC, BRAF, MAP2K4, RB1, STAT4, STAT1, and RET.
In another aspect, the invention is directed to a method of determining a TNBC LAR subtype in an individual in need thereof comprising determining increased expression of one or more hormone regulated genes in the TNBC cells compared to the control. Increased expression of the one or more genes indicates that the TNBC is a TNBC LAR type. In one aspect, the method of determining a TNBC LAR subtype in an individual in need thereof comprises selectively detecting increased expression of a gene combination comprising AR, DHCR24, ALCAM, GATA2, GATA3, IDIH1, IDIH2, CDH11, ERBB3, CUX2, FGFR4, HOPX, FASN, FKBP5, APOD, PIP, SPDEF, CLDN8, FOXA1, KRT18, and XBP1 wherein detection of increased expression of the gene combination indicates that the TNBC is TNBC LAR subtype. Determination that the TNBC is a LAR subtype TNBC can further comprise detecting one or more mutated genes. Examples of such mutated genes include PIK3CA, CDH1, PTEN, RB1, TP53, and MAP3K1.
A variety of methods (e.g. spectroscopy, colorometry, electrophoresis, chromatography) can be used to detect increased expression of a (one or more) gene. As is apparent to those of skill in the art, increased expression of one or more genes can be detected by measuring all or a portion of nucleic acid (e.g., DNA, mRNA) of the gene and/or protein (polypeptide) expressed by the gene. The level, expression and/or activity of a gene and or its encoded polypeptide can be measured. For example, nucleic acid such as genes, mRNA or a combination thereof, can be determined using PCR (e.g., RT-PCR, RT-qPCR), gene chips or microarray analysis (DNA arrays, gene expression arrays, nanostrings), next generation sequencing (e.g., next generation RNA sequencing), in situ hybridization, blotting techniques, and the like. Protein can be detected using immunoassays (e.g., immunohistochemistry, immunofluorescence, immunoprecipitation), arrays (e.g., reverse phase protein microarrays), blotting techniques (e.g., Western blots), SDS-PAGE, and the like
As described herein, the methods can further comprise detecting one or more mutated genes. A variety of methods can be used to detect one or more mutated genes. Examples of such methods include a SNaPshot assay, exome sequencing, Sanger gene sequencing, resequencing array analysis, mRNA analysis/cDNA sequencing polymerase chain reaction (PCR), single-strand conformation polymorphism (sscp), heteroduplex analysis (het), allele-specific oligonucleotide (aso), restriction fragment analysis, allele-specific amplification (asa), single nucleotide primer extension, oligonucleotide ligation assay (ola), denaturing gradient gel electrophoresis (DGGE), temperature gradient gel electrophoresis (TGGE) and single strand conformation polymorphism (SSCP).
The methods can further comprise comparing the expression of the one or more genes in the individual to the expression of the one or more genes in a control. As will be apparent to those of skill in the art, a variety of suitable controls can be used. Examples of such controls include one or more non-cancer cells (e.g., breast cells from one or more individuals that do not have cancer) and/or non-TNBC cells (e.g., from the individual being tested). The control can also be a (one or more) standard or reference control established by assaying one or more (e.g., a large sample of) individuals which do not have cancer (e.g., TNBC) and using a statistical model to obtain a control value (standard value; known standard). See, for example, models described in Knapp, R. G. and Miller M. C. (1992) Clinical Epidemiology and Biostatistics, William and Wilkins, Harual Publishing Co. Malvern, PA, which is incorporated herein by reference.
As used herein an “individual” refers to an animal, and in a particular aspect, a mammal. Examples of mammals include primates, a canine, a feline, a rodent, and the like. Specific examples include humans, dogs, cats, horses, cows, sheep, goats, rabbits, guinea pigs, rats and mice.
The term “individual in need thereof” refers to an individual who is in need of treatment or prophylaxis as determined by a researcher, veterinarian, medical doctor or other clinician. In one aspect, an individual in need thereof is a mammal, such as a human. In other aspects, the individual has been diagnosed with breast cancer, the individual has not been diagnosed with breast cancer, or the individual is at risk of developing breast cancer.
The methods of the invention can further comprise detecting expression (increased expression) of one or more genes in a sample (biological sample) of the individual. Thus, the method can further comprise obtaining a (one or more) sample from the individual. The sample can be, for example, a biological fluid, a biological tissue (e.g., a biopsy) and combinations thereof from the individual. In particular aspects, the sample is one or more TNBC cells (e.g., epithelial cells, mesenchymal cells, immune cells). Methods for obtaining such biological samples from an individual are known to those of skill in the art.
As also shown herein, the methods can further comprise determining a treatment protocol for the triple negative breast cancer (TNBC) patient based on the TNBC subtype. As described herein, a “cancer therapy”, “treatment for cancer” or “cancer treatment” comprises administering one or more agents to the individual. In one embodiment, the agent is a chemotherapeutic agent which targets the TNBC cells. Examples of such agents include an alkylating agent (e.g., busulfan, cisplatin, carboplatin, chlorambucil, cyclophosphamide, ifosfamide, dacarbazine (DTIC), mechlorethamine (nitrogen mustard), melphalan, temozolomide); a nitrosoureas (e.g., carmustine (BCNU), lomustine (CCNU)); an antimetabolite (e.g., 5-fluorouracil, capecitabine, 6-mercaptopurine, methotrexate, gemcitabine, cytarabine (ara-C), fludarabine, pemetrexed); an anthracycline or related drug (e.g., daunorubicin, doxorubicin (adriamycin), epirubicin, idarubicin, mitoxatrone); a topoisomerase (topoisomerase I inhibitor; topoisomerase inhibitor II) inhibitor (e.g., topotecan, irinotecan, etoposide (VP-16), teniposide); and a mitotic inhibitor (e.g., taxanes (paclitaxel, docetaxel, taxol) vinca alkaloids (vinblastine, vincristine, vinorelbine).
In another embodiment, the agent is a targeted therapy agent which attacks cancer cells more specifically than chemotherapeutic agents. Examples of such agents include imatinib (Gleevec), gefitinib (Iressa), erlotinib (Tarceva), rituximab (Rittman), and bevacizumab (Avastin). In yet another embodiment, the agent is a sex hormone, or hormone-like drug that alters the action or production of female or male hormones and can be used to slow the growth of breast, prostate, and endometrial (uterine) cancers. Examples of such agents include anti-estrogens (e.g., tamoxifen, fulvestrant), aromatase inhibitors (e.g., anastrozole, exemestane, letrozole), progestins (e.g., megestrol acetate), anti-androgens (e.g., bicalutamide, flutamide) and LHRH agonists (leuprolide, goserelin). In addition, the agent can be a drug which is used to stimulate the immune system to more effectively recognize and attack cancer cells of an individual with cancer. Other examples of agents include a hormone such as a selective estrogen receptor modulator (SERM); an antibody or antigen binding fragment thereof (e.g., herceptin); a protein tyrosine kinase inhibitor; a combination of chemotherapeutic agents such as cytoxan (C), methotrexate (M), fluorouracil (F), an anthracylcin such as adriamycin (A), epirubicin (E), doxorubicin, and/or daunorubicin; a targeted kinase inhibitor; a metalloproteinase inhibitor; and a proteosome inhibitor.
The one or more agents can be administered to the individual before (e.g., neoadjuvant therapy), during or after (e.g., adjuvant therapy) primary treatment of the cancer. As used herein “primary treatment of a cancer” generally refers to a treatment involving surgery (e.g., surgical removal of a tumor) and/or radiation (e.g., local radiation). In the methods of the present invention, the “treatment for cancer” can be a neoadjuvant treatment wherein an agent (e.g., a hormone, a chemotherapeutic) is administered prior to surgical removal of a (residual) tumor, or prior to localized radiation (e.g., radiation used to shrink the tumor in order to simplify subsequent surgical removal of the tumor). In another embodiment, the “treatment for cancer” can be an adjuvant treatment wherein an agent (e.g., a hormone, a chemotherapeutic agent) is administered after surgical removal of a (residual) tumor, or after localized radiation. In yet another embodiment, the treatment can be a palliative treatment in which one or more chemotherapeutic agents are administered to an individual to treat metastatic cancer (e.g., to make the individual more comfortable and/or to prolong survival of the individual after the cancer has metastasized).
In particular aspects, the TNBC BL1 or BL2 subtype is detected in the individual and the individual is treated with one or more drugs that damages DNA. Examples of such drugs include an alkylating-like agent (e.g., cisplatin) and a PARP inhibitor (e.g., veliparib, olaparib).
In another aspect, the TNBC ML subtype is detected in the individual and the individual is treated with one or more drugs that inhibits Src (e.g., disatinib), IGF1R (e.g., OSI-906, BMS-754807), MET (e.g., PF2341066), PDGFR (e.g., sorafinib), P13K (e.g., BKM-120, GDC0941), P13K/mTOR (e.g., NVP-BEZ235, GDC0980) or a combination thereof.
In another aspect, the TNBC LAR subtype is detected in the individual and the individual is treated with one or more drugs that inhibit androgen receptor (AR). Examples of such drugs include bicalutamide, MVD3100, and abiraterone. The method can further comprise treating the individual with P13K/mTOR (e.g., NVP-BEZ235, GDC0980) or a combination thereof. The method can further comprise treating the individual with an inhibitor of HSP90 (e.g., DMAG).
In other aspects, the invention can further comprise predicting whether an individual (e.g., patient) will benefit from a (one or more) treatment for a particular TNBC subtype.
Also described herein are TNBC cell lines that serve as models for each of the six TNBC subtypes. Thus, in another aspect, the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) subtype comprising contacting a cell line that is a model for the TNBC subtype of interest with an agent to be assessed; and determining viability of the cell line in the presence of the agent, wherein if the viability of the cell line decreases in the presence of the agent, then the agent can be used to treat the TNBC subtype of interest. The TNBC subtype of interest can be a TNBC BL-1 subtype, a TNBC BL-2 subtype, a TNBC IM subtype, a TNBC M subtype, a TNBC MSL subtype or a TNBC LAR subtype. In addition, as will be apparent to those of skill in the art, the viability of a cell decreases when e.g., the cell undergoes apoptosis, proliferation of the cell is inhibited or slowed and/or metastasis of the cell is inhibited to slowed.
Examples of agents (test agent; agent of interest) include nucleic acids, polypeptides, fusion proteins, peptidomimetics, prodrugs, drugs, receptors, binding agents (e.g., antibodies), small molecules, etc and libraries (e.g., combinatorial libraries) of such agents. Test agents can be obtained made or obtained from libraries of natural, synthetic, and/or semi-synthetic products (e.g., extracts). Those skilled in the field of drug discovery and development will understand the precise source of such agents.
In one aspect, the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) BL-1 subtype comprising contacting one or more TNBC BL-1 subtype cell lines with an agent to be assessed. The viability of the one or more cell lines in the presence of the agent is detected, wherein if the viability of the one or more cell lines decreases in the presence of the agent, then the agent can be used to treat the TNBC BL-1 subtype. Examples of TNBC BL-1 subtype cell lines include HCC2157, HCC1599, HCC1937, HCC1143, HCC3153, MDA-MB-468, and HCC38.
In another aspect, the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) BL-2 subtype comprising contacting one or more TNBC BL-2 subtype cell lines with an agent to be assessed. The viability of the one or more cell lines in the presence of the agent is determined, wherein if the viability of the one or more cell lines decreases in the presence of the agent, then the agent can be used to treat the TNBC BL-2 subtype. Examples of TNBC BL-2 subtype cell lines include SUM149PT, CAL-851, HCC70, HCC1806 and HDQ-P1.
In another aspect, the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) IM subtype comprising contacting one or more TNBC IM subtype cell lines with an agent to be assessed. The viability of the one or more cell lines in the presence of the agent is determined, wherein if the viability of the one or more cell lines decreases in the presence of the agent, then the agent can be used to treat the TNBC IM subtype. Examples of TNBC IM subtype cell lines include HCC1187 and DU4475.
In another aspect, the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) M subtype comprising contacting one or more TNBC M subtype cell lines with an agent to be assessed. The viability of the one or more cell lines in the presence of the agent is determined, wherein if the viability of the one or more cell lines decreases in the presence of the agent, then the agent can be used to treat the TNBC M subtype. Examples of TNBC M subtype cell lines include BT-549, CAL-51 and CAL-120.
In another aspect, the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) MSL subtype comprising contacting one or more TNBC MSL subtype cell lines with an agent to be assessed. The viability of the one or more cell lines in the presence of the agent is determined, wherein if the viability of the one or more cell lines decreases in the presence of the agent, then the agent can be used to treat the TNBC MSL subtype. Examples of TNBC MSL subtype cell lines include HS578T, MDA-MB-157, SUM159PT, MDA-MB-436, and MDA-MB-231.
In another aspect, the invention is directed to a method of determining whether an agent can be used to treat a triple negative breast cancer (TNBC) LAR subtype comprising contacting one or more TNBC LAR subtype cell lines with an agent to be assessed. The viability of the one or more cell lines in the presence of the agent is determined, wherein if the viability of the one or more cell lines decreases in the presence of the agent, then the agent can be used to treat the TNBC LAR subtype. Examples of TNBC LAR subtype cell lines include MDA-MB, SUM185PE, HCC2185, CAL-148, and MFM-223.
In the methods of the invention, viability of one of more of the cells can be determined using a variety of methods. Examples of such methods include a membrane leakage assay, a mitochondrial activity assay, a functional assay, a proteomic assay, a genomic assay or a combination thereof.
The methods can further comprise comparing the viability of the one or more cell lines in the presence of the agent to the viability of a control such as a non-cancerous cell line (e.g., a culture of normal human and immortalized MCF210A).
EXEMPLIFICATION Example 1 Identification of Human Triple Negative Breast Cancer Subtypes and Models for Selection of Targeted Therapies
Methods
Laser capture microdissection, RNA extraction, and GE profiling. Invasive tumor cells from serial sections of primary breast cancers (n=112) were captured onto polymeric caps using the PixCell II lasercapture microdissection system (Arcturus) as previously described (Bauer J A, et al. Clin Cancer Res. 2010; 16(2):681-690). Areas of ductal carcinoma in situ and normal breast tissue were excluded and areas of inflammation and areas with tumor-associated fibroblasts were avoided. Total RNA was isolated from captured cells, quantified, and integrity analyzed, and microarray analyses were done using Affymetrix GeneChip Human Genome U133 Plus 2.0 arrays as previously described (Bauer J A, et al. Clin Cancer Res. 2010; 16(2):681-690).
Data set collection and TNBC identification by bimodal filtering. 2353 human breast cancer GE profiles were compiled from 14 publicly available breast cancer microarray data sets (GEO, www.ncbi.nlm.nih.gov/gds; Array Express, www.ebi.ac.uk/microarray-as/ae/) including 112 primary breast cancer GE profiles from our institution (Vanderbilt University Medical Center). All Vanderbilt tissue samples were taken from individuals treated at Vanderbilt University with institutional review board approval, and all patients signed a protocol-specific consent. All GE profiles were generated on Affymetrix microarrays and collected for identification of TNBCs for the training data set (Table 1). An additional 894 breast cancer GE profiles from 7 data sets were collected for the identification of TNBCs for the validation data set (Table 2). Raw GE values for each data set (n=21) were normalized independently using RMA procedure. The Affymetrix probes 205225_at, 208305_at and 216836_s_at were chosen to represent ER, PR, and HER2 expression, respectively (Karn T, et al. Breast Cancer Res Treat. 2010; 120(3):567-579). For each data set, empirical expression distributions of ER, PR, and HER2 were analyzed using a 2-component Gaussian mixture distribution model and parameters were estimated by maximum likelihood optimization, using optim function (R statistical software, www.r-project.org). The posterior probability of negative expression status for ER, PR, and HER2 was then estimated. A sample was classified as having negative expression if its posterior probability was less than 0.5. Upon initial hierarchical clustering, it was evident that some samples were from the same tumors, but in different data sets. These and additional outliers were removed after PCA. To ensure all ER/PR/HER2-positive tumors were removed, a secondary filter was performed on the combined samples identified by bimodal filtering. TNBC tumors were renormalized along with 5 positive controls for each parameter (ER, PR, and HER2) from 4 data sets that were positive by IHC and expressed mRNA near the center of the positive bimodal peak (FIG. 1B). Only tumors that displayed a greater than 10-fold reduction in expression than the positive controls were considered negative in expression and used for further analysis, resulting in 386 TNBC tumors (training set) and 201 TNBC tumors (validation set).
Normalization, data reduction, and cross-platform batch effect removal. Training (n=386) and validation (n=201) TNBC data sets identified by the 2-component Gaussian distribution model were collectively RMA normalized, summarized and log-transformed using the combined raw GE from each data set. For genes containing multiple probes, the probe with the largest interquartile range across the samples was chosen to represent the gene. Batch effects were removed by fitting each gene to a linear model with 14 and 7 fixed effects for each data set, respectively. The residual genes from this model (n=13,060) were used for subsequent clustering analysis.
GE-based identification of TNBC subtypes. k-means clustering and consensus clustering were used to determine the optimal number of stable TNBC subtypes. Cluster robustness was assessed by consensus clustering using agglomerative k-means clustering (1,000 iterations), with average linkage on the 386 TNBC profiles using the most differentially expressed genes (SD >0.8; n=1510 genes) (Gene Pattern version 3.2.1, GSE-A, www.broadinstitute.org/gsea/) (19). The optimal number of clusters was determined from the CDF, which plots the corresponding empirical cumulative distribution, defined over the range [0, 1], and from calculation of the proportion increase in the area under the CDF curve. The number of clusters is decided when any further increase in cluster number (k) does not lead to a corresponding marked increase in the CDF area. PCA and heat maps were generated using Genespring GX ver.10 software (Agilent).
Functional annotation. Each TNBC subtype was tested for gene enrichment compared with all other samples using GSE-A software (Subramanian A, et al. Proc Natl Acad Sci USA. 2005; 102(43):15545-15550). Genes were tested for enrichment in the C2 curated gene sets of canonical pathways. Using the GSE-A algorithm (1,000 permutations), the top significantly enriched canonical pathways were selected based on a normalized enrichment score (NES) greater than 0.4 and false discovery rate (FDR) q value of less than 0.60. The FDR cutoff of 0.60 was chosen because of the lack of coherence in the data set collection spanning 21 studies, and more stringent FDR cutoffs resulted in fewer results, potentially overlooking significant pathways.
TNBC subtype gene signature derivation. The 20% of genes with the highest and lowest expression levels in at least 50% of the samples for each subtype were chosen for further analysis. Within each cluster, for each selected gene, the nonparametric Kruskal-Wallis test was applied to identify genes significantly different from the median GE among all 6 groups. Significant genes with a Bonferroni's adjusted P value of less than 0.05 were included in the combined gene signature (n=2188) for the TNBC subtypes and used to predict an independent validation set as well as to classify TNBC cell lines. Each sample from the validation set was assigned to a TNBC subtype based on highest Pearson correlation and lowest P value to one of the subtypes derived from the training data set. Samples with correlations differing by a P value of less than 0.05 were considered unclassifiable.
TNBC cell-line classification. GE data for TNBC cell lines (GSE-10890 and E-TABM-157) were correlated (Spearman) to the centroids of the GE signatures for each TNBC subtype. GE data from both the TNBC tumors and cell lines were combined so that each gene was standardized to have mean=0 and SD=1. GE profiles from the cell lines were correlated to the centroids for each of the 6 TNBC subtypes. To remove size effects of the 6 gene signatures, the empirical P values were estimated for the correlations using a resampling (bootstrap, n=1000) approach and estimating correlation coefficients for each resample. Cell lines were assigned to the TNBC subtype with the highest correlation (Table 3), and those that had low correlations (<0.1) or were similar between multiple subtypes (P>0.05) were considered unclassified.
Cell proliferation/viability assays and IC50 determinations. All cell lines and culture conditions used can be found in Supplemental Methods (Table 5). Short tandem repeat (STR) DNA fingerprinting analysis was performed on all TNBC cell lines used in this study (Cell Line Genetics). All cell lines matched STR databases (ATCC, DSMZ, and Asterand), and their identity was confirmed by a certified process at Cell Line Genetics. Breast cancer cell lines and HMECs were seeded (3,000-10,000 cells) in quadruplicate wells in 96-well plates. Cells were incubated in the presence of alamar-Blue (Invitrogen), and baseline (predrug) fluorescence (Ex/Em: 560/590 nm) measurements were obtained following overnight attachment. Medium was then replaced with either fresh medium (control) or medium containing half-log serial dilutions of the following drugs: 0.3-30 μM olaparib (ChemieTek), 0.3-30 μM veliparib (ChemieTek), 1-100 μM bicalutamide (IPR Pharmaceutical), 0.3-30 μM cisplatin (APP Pharmaceutical), 3-300 nM NVP-BEZ235 (Chemdea), 10-1000 nM 17-DMAG (ChemieTek), and 0.1-10 μM dasatinib (LC Laboratories). Viability was determined from measuring fluorescent intensity after metabolic reduction of alamarBlue in the presence/absence of the drug after 72 hours. Viability assays were performed in triplicate, and replicates were normalized to untreated wells. Inhibitory concentration (ICH) values were determined after double-log transformation of dose response curves as previously described (Bauer J A, et al. Breast Cancer Res. 2010; 12(3):R41). For analysis of cell-line drug assays, data generated from the different cell lines representative of TNBC subtypes were compared using the nonparametric Mann-Whitney U test. All analyses and graphic representations were performed using Prism software (version 4.0c; GraphPad Software), and values are represented as the mean±SEM.
Kaplan-Meier survival analysis and Hr. Log-rank test was used to determine survival significance in TNBC subtypes from Kaplan-Meier survival curves, RFS, and DMFS. Cox proportional hazards model was used to calculate Hr, demonstrating differences in survival by pairwise comparison between TNBC subtypes (P<0.05).
Xenograft tumor studies. Five-week-old female athymic nude-Foxnlnu mice (Harlan Sprague-Dawley) were injected (s.c.) with either approximately 5×106 (CAL-51, HCC1806, MDA-MB-468, and SUM185PE) or approximately 10×106 (CAL-148 and SUM159PT) cells suspended in medium (200 μl) into each flank using a 22-gauge needle. The protocols describing the process of xenograft tumor formation in athymic mice were reviewed and approved by the Vanderbilt Institutional Review Board. Once tumors reached a volume of 25-50 mm3, mice were randomly allocated to treatment or vehicle arms. Treatments included bicalutamide per oral (100 mg/kg/d), cisplatin i.p. (8 mg/kg/wk), or NVP-BEZ235 per oral (50 mg/kg/d) in 1:9 N-methylpyrolidone: PEG300. Tumor diameters were serially measured at the indicated times with digital calipers, and tumor volumes were calculated as width2×length/2. Data points reflect the means and SEM for 16 tumors per treatment.
Statistics. Description of the relevant statistical methods used and analyses performed are described in the relevant Methods sections above. Comparisons between cell lines representative of TNBC subtypes were performed using the nonparametric Mann-Whitney U test. Statistical significance of drug effects on tumor growth in athymic mice was determined by 2-tailed unpaired t test.
Supplemental Methods
Immunostaining. Both formalin fixed paraffin embedded (FFPE) and frozen tissue were used for immunohistochemical studies. When frozen tissue was used for immunohistochemistry, staining was performed on sections taken from the same tissue block from which RNA was isolated for microarray. AR, Ki67, EGFR and CK 5/6 expression were evaluated in frozen tissue using the DAKO (Carpinteria, CA) antibodies: AR (clone AR411) at a 1:50 dilution, EGFR at 1:200, CK5/6 at 1:50 and Ki67 antibody (MIB1 clone) at a 1:200 dilution for 1 h at room temperature. FFPE tissue was subject to antigen retrieval with high pH buffer (pH 8.0) followed by overnight incubation with an AR (1:30) or Ki67 (1:75) antibody dilution overnight. AR expression was scored as both the percentage of tumor cells with nuclear staining as well as the intensity of staining (scored as 0-3+). An AR intensity score was calculated as follows: (AR intensity×100)+% AR positive cells. For Ki67 the percentage of cells demonstrating nuclear staining at any intensity was recorded.
Immunoblotting. Cells were trypsinized, lysed and relative protein expression was determined by Western blot as previously described with the following antibodies; HSP90 monoclonal antibody, clone F-18 (Santa Cruz Biotechnology, Santa Cruz, CA) and the AR polyclonal antibody, SC-N20 (Santa Cruz Biotechnology).
Colony Formation. MFM-223, MDA-MB-453 and CAL-148 cells (3000 cells/well) were reverse-transfected with 1.25 pmole of siRNAs to AR [ONTARGET plus SMARTpool, cat #L-003400-00 (Dharmacon, Lafayette, CO)] or non-targeting control (ON-TARGETplus Non-targeting Pool cat #D-001810-10-05) with 0.25 μL Dharmafect #3 (MFM-223), or 0.25 μL Dharmafect #1 (MDAMB-453 and CAL-148). Colonies were stained and quantified 14 d following transfection using Cell Profiler 2.0 (Broad Institute, Cambridge, MA). Experiments were preformed in triplicate and error bars reflect standard deviation.
Results
Analysis of human tumor GE profiles identifies TNBC subtypes. GE profiles were obtained from 21 publicly available data sets that contained 3,247 primary human breast cancers and processed according to the flow chart in FIG. 1A. To allow for robust analysis, these data sets were further divided into training (Table 1; 14 data sets, cases n=2353) and validation data sets (Table 2; 7 data sets, cases n=894). Since the majority of these tumors lacked sufficient molecular analysis of ER, PR, and HER2, we filtered each data set for ER, PR, and HER2 mRNA expression to identify triple-negative status (see Methods for bimodal filter description; supplemental material available in Lehmann, B. D., et al., J Clin Invest, 121 (7): 2750-2767 (July 2011), the entire teachings of which are incorporated herein by reference). Previous studies have shown that ER and HER2 mRNA expression correlates with immunohistochemistry (IHC) and FISH analyses, respectively (Carmeci C. et al. Am J Pathol. 1997; 150(5):1563-1570; Press M F, et al. Clin Cancer Res. 2008; 14(23):7861-7870). Using a bimodal filter on the GE data (FIG. 1B), 386 and 201 TNBC tumor samples were identified in the training and validation sets, respectively. The 386 TNBC GE profiles of the training set were robust multiarray average (RMA) normalized, summarized, transformed, and corrected for “batch effect,” resulting in 13,060 identical probes representing unique genes across all platforms. The triple-negative GE pattern is shown compared with 5 positive controls for each parameter (ER, PR, and HER2) from data sets that were confirmed IHC positive and expressed mRNA near the center of the positive bimodal peak (FIG. 1C). Of the 14 data sets in the training set, 4 (GSE-7904, E-TABM-158, MDA133, GSE-22513, GSE-28821, and GSE-28796) included IHC data for all 3 markers, while others lacked information on ER, PR, or HER2 status (Table 1). The IHC data provided were used to calculate false-positive rates for each study, defined as tumors that were predicted negative for ER, PR, or HER2 by bimodal filtering of mRNA expression, but were positive by IHC. The overall false-positive rates were 1.7%, 1.7%, and 0.9% for ER, PR, and HER2, respectively, demonstrating that bimodal filtering of data sets by mRNA expression is a reliable method for identifying TNBC tumors from data sets lacking IHC information (Table 1). The overall frequency of TNBC across the training data set was 16% and is consistent with the prevalence of TNBC previously reported in 2 other large studies performed on 3,744 cases (17%) (17) and 1,726 cases (16%) (18).
k-means and consensus clustering reveal 6 TNBC subtypes. To identify global differences in GE between TNBC subtypes, k-means clustering was performed on the most differentially expressed genes (SD >0.8). Using the silhouette width (s[i]) as a measure of relative closeness of individual samples to their clusters, k-means clustering classified 337 of the 386 TNBC tumors into 6 stable clusters (s[i]>0) and 49 tumors into 1 unstable cluster (s[i]>0) (FIG. 2A). Clustering resulted in a distribution of samples in all 7 clusters independent of each data set, n=14, indicating that confounding factors such as batch effect, RNA amplification, and sample quality did not influence cluster distribution (FIG. 8 ). Sample classification robustness was analyzed by consensus clustering, which involves k-means clustering by resampling (1,000 iterations) randomly selected tumor profiles. The consensus matrix is a visual representation of the proportion of times in which 2 samples are clustered together across the resampling iterations (FIG. 2B). Groups of samples that frequently cluster with one another are pictorially represented by darker shades of red. To determine the number of clusters present in the data, the area under the curve of the consensus distribution function (CDF) plot was examined (FIG. 2C). The point at which the area under the curve ceases to show marked increases with additional cluster number (k) indicates the ideal number of clusters (FIG. 2D; Monti S. Machine Learning. 2003; 52(1-2):91-118). Therefore, the optimal number of clusters is 7, as defined by the consensus plots consistent with the k-means clustering (6 stable, 1 unstable). Unsupervised dimension reduction by principal component analysis demonstrated fundamental differences in GE between tumor subtypes identified by k-means and consensus clustering (FIG. 2E).
To assess whether similar TNBC subtypes could be generated from an independent TNBC cohort, 201 TNBC samples were compiled from bimodal filtering of 7 additional publicly available data sets (Table 2). A GE signature was derived from the most differentially expressed genes found in the TNBC training set (see Methods) and used to predict which TNBC subtype was best-fit for each of the tumors in the validation set. Each sample from the validation set was assigned to 1 of the TNBC subtypes derived from the training data set based on the highest Pearson correlation and lowest P value. Samples with correlations differing by P<0.05 were considered unclassifiable. The validation set resulted in proportioned subtypes similar to those of the initial k-means clustering of the training set. After the analysis was completed, an ad hoc analysis was performed by combining the training and validation data sets (587 tumors), which resulted in 7 subtypes identified by consensus clustering (FIG. 9 ), with similar enrichment in gene ontologies. This further validates the stability of the subtypes and shows that increasing sample size does not change optimal cluster number. Evaluation of GE profiles of RNA obtained from laser-capture microdissection of tumor cells from 2 data sets (GSE-22513; GSE-28821; GES-28796, Table 1; GSE-5847, Table 2) showed a similar pattern of distribution across all 7 subtypes, suggesting that these subtypes are indeed representative primarily of GE resulting from epithelial tumor cells rather than stromal components surrounding the tumor (i.e., inflammatory cells, myofibroblasts, etc.) (FIG. 10 ).
Distinct gene ontologies are associated with each TNBC subtype. As an independent method to analyze TNBC subtypes, Gene Set Enrichment Analysis (GSE-A) (Subramanian A, et al. Proc Natl Acad Sci USA. 2005; 102(43):15545-15550) was performed on all genes from both the training and validation sets to determine the top canonical pathways associated with each TNBC subtype. The top enriched genes (P<0.05), identified from the training set used to predict the validation set, are displayed in a heat map in FIG. 3 . The top canonical pathways substantially overlapped between the training and validation sets for each subtype, indicating that the subtypes were reproducibly enriched in unique pathways (Table 4). Our 7 TNBC subtypes were characterized on the basis of gene ontologies and differential GE and subsequently labeled as follows: basal-like 1 (BL1); basal-like 2 (BL2); immunomodulatory (IM); mesenchymal (M); mesenchymal stem-like (MSL); luminal androgen receptor (LAR); and unstable (UNS) (FIG. 3 ). Independent analysis of 5 data sets based on TNBCs identified by IHC staining (n=183) resulted in 4 clusters with GE similar to that of basal-like, IM, mesenchymal-like, and LAR subtypes (FIGS. 11A, 11B and 12 ).
BL1 and BL2 subtypes. The top gene ontologies for the BL1 subtype are heavily enriched in cell cycle and cell division components and pathways (cell cycle, DNA replication reactome, G2 cell-cycle pathway, RNA polymerase, and G1 to S cell cycle) (FIG. 3 ). The annotations are supported by the expression of genes associated with proliferation, such as AURKA, AURKB, CENPA, CENPF, BUB1, TTK, CCNA2, PRC1, MYC, NRAS, PLK1, and BIRC5 (FIG. 13 ). Elevated DNA damage response (ATR/BRCA) pathways accompany the proliferation pathways in the BL1 subtype (FIG. 3 ). Increased proliferation and cell-cycle checkpoint loss are consistent with the elevated expression of the DNA damage response genes observed (CHEK1, FANCA, FANCG, RAD54BP, RAD51, NBN, EXO1, MSH2, MCM10, RAD21, and MDC1) (FIG. 13 ).
The highly proliferative nature of this subtype is further supported by the finding of high Ki-67 mRNA expression (MKI67) (FIG. 13 ) and nuclear Ki-67 staining as assessed by IHC analysis (BL1+BL2=70% vs. other subtypes=42%; P<0.05) (FIGS. 14A-14B). Enrichment of proliferation genes and increased Ki-67 expression in basal-like TNBC tumors suggest that this subtype would preferentially respond to antimitotic agents such as taxanes (paclitaxel or docetaxel) (Chakravarthy A B, et al. Clin Cancer Res. 2006; 12(5):1570-1576; Bauer J A, et al. Clin Cancer Res. 2010; 16(2):681-690). This is indeed the case when comparing the percentage of patients achieving a pathologic complete response (pCR) in 42 TNBC patients treated with neoadjuvant taxane in 2 studies (Bauer J A, et al. Clin Cancer Res. 2010; 16(2):681-690; Juul N, et al. Lancet Oncol. 2010; 11(4):358-365). In these combined studies, TNBC patients whose tumors correlated to the basal-like (BL1 and BL2) subtype had a significantly higher pCR (63%; P=0.042) when treated with taxane-based therapies as compared with mesenchymal-like (31%) or LAR (14%) subtypes (FIG. 15 ).
The BL2 subtype displays unique gene ontologies involving growth factor signaling (EGF pathway, NGF pathway, MET pathway, Wnt/β-catenin, and IGF1R pathway) as well as glycolysis and gluconeogenesis (FIG. 3 ). Likewise, the BL2 subtype is uniquely enriched in growth factor receptor GE such as EGFR, MET, and EPHA2 (FIG. 13 ). This subtype has features suggestive of basal/myoepithelial origin as demonstrated by higher expression levels of TP63 and MME (CD10) (FIG. 13 ).
IM subtype. The IM subtype is enriched for gene ontologies in immune cell processes. These processes include immune cell signaling (TH1/TH2 pathway, NK cell pathway, B cell receptor [BCR] signaling pathway, DC pathway, and T cell receptor signaling), cytokine signaling (cytokine pathway, IL-12 pathway, and IL-7 pathway), antigen processing and presentation, and signaling through core immune signal transduction pathways (NFKB, TNF, and JAK/STAT signaling) (FIG. 3 ). The IM signaling is evidenced by immune signal transduction GE (FIG. 13 ), in addition to immune cell-surface antigens, cytokine signaling, complement cascade, chemokine receptors and ligands, and antigen presentation (FIG. 16 ). Since a similar proportion of samples that were microdissected fell into the IM subtype, it is likely that the IM characteristics are unique to the tumor cells themselves and not a reflection of immune cell infiltrate (FIG. 10 ). Immune signaling genes within the IM subtype substantially overlap with a gene signature for medullary breast cancer, a rare, histologically distinct form of TNBC that despite its high-grade histology is associated with a favorable prognosis (Bertucci F, et al. Cancer Res. 2006; 66(9):4636-4644).
M and MSL subtypes. The M subtype displays a variety of unique gene ontologies that are heavily enriched in components and pathways involved in cell motility (regulation of actin by Rho), ECM receptor interaction, and cell differentiation pathways (Wnt pathway, anaplastic lymphoma kinase [ALK] pathway, and TGF-β signaling) (FIG. 3 ). The MSL subtype shares enrichment of genes for similar biological processes with the M subtype, including cell motility (Rho pathway), cellular differentiation, and growth pathways (ALK pathway, TGF-β signaling and Wnt/β-catenin pathway). However, unique to the MSL are genes representing components and processes linked to growth factor signaling pathways that include inositol phosphate metabolism, EGFR, PDGF, calcium signaling, G-protein coupled receptor, and ERK1/2 signaling as well as ABC transporter and adipocytokine signaling (FIG. 3 ).
The prevalence of cell differentiation and growth factor signaling pathways is illustrated by expression of TGF-β signaling pathway components (TGFB1L1, BGN SMAD6, SMAD7, NOTCH1, TGFB1, TGFB2, TGFB3, TGFBR1, TGFBR2, and TGFBR3), epithelial-mesenchymal transition-associated (EMT-associated) genes (MMP2, ACTA2, SNAI2, SPARC, TAGLN, TCF4, TWIST1, ZEB1, COL3A1, COL5A2, GNG11, ZEB2, and decreased E-cadherin [CDH1] expression), growth factors (FGF, IGF, and PDGF pathways), and Wnt/β-catenin signaling (CTNNB1, DKK2, DKK3, SFRP4, TCF4, TCF7L2, FZD4, CAV1, CAV2, and CCND2) (FIG. 13 ). The MSL subtype is also uniquely enriched in genes involved in angiogenesis, including VEGFR2 (KDR), TEK, TIE1, and EPAS1 as well as immune signaling evidenced by an overlap in GE unique to the IM subtype (FIG. 16 ).
One interesting difference between the M and MSL subtypes is that the MSL subtype expresses low levels of proliferation genes (FIG. 13 ). This decreased proliferation is accompanied by enrichment in the expression of genes associated with stem cells (ABCA8, PROCR, ENG, ALDHA1, PER1, ABCB1, TERF2IP BCL2, BMP2, and THY1), numerous HOX genes (HOXA5, HOXA10, MEIS1, MEIS2, MEOX1, MEOX2, and MSX1), and mesenchymal stem cell-specific markers (BMP2, ENG, ITGAV, KDR, NGFR, NT5E, PDGFRB, THY1, and VCAM1) (FIG. 13 ). The signaling pathway components differentially expressed in the M and MSL groups share similar features with a highly dedifferentiated type of breast cancer called metaplastic breast cancer, which is characterized by mesenchymal/sarcomatoid or squamous features and is chemoresistant (Gibson G R, et al. Am Surg. 2005; 71(9):725-730).
The MSL subtype also displays low expression of claudins 3, 4, and 7, consistent with a recently identified claudin-low subtype of breast cancer (FIGS. 13 and 17 ; Prat et al., Breast Cancer Res, 2010; 12(5):R68). Hierarchical clustering of TNBC GE profiles using the claudin-low gene predictor set (n=770) segregated a portion of the M and MSL subtypes with low claudin (3, 4, and 7), cytokeratin (KRT7, KRT8, KRT18, and KRT19), and CD24 expression (FIG. 17 ; Prat A, et al. Breast Cancer Res. 2010; 12(5):R68). This population of claudin-low-expressing tumors also had high expression of genes associated with EMT (FBN1, MMP2, PDGFRB, THY1, SPARC, TGFBR2, PDGFRA, TWIST, CAV1, CAV2, and SERPINE1).
LAR subtype. GE in the LAR group was the most differential among TNBC subtypes. This subtype is ER negative, but gene ontologies are heavily enriched in hormonally regulated pathways including steroid synthesis, porphyrin metabolism, and androgen/estrogen metabolism (FIG. 3 ). Whether other hormone-regulated pathways such as androgen receptor (AR) signaling, previously reported in ER-negative breast cancer (Hayes M J et al. Clin Cancer Res. 2008; 14(13):4038-4044), could be responsible for the GE patterns in this LAR subtype was investigated. Indeed, it was found that AR mRNA was highly expressed, on average at 9-fold greater than all other subtypes (FIG. 13 ). Tumors within the LAR group also expressed numerous downstream AR targets and coactivators (DHCR24, ALCAM, FASN, FKBP5, APOD, PIP, SPDEF, and CLDN8) (FIG. 13 ). In addition to AR mRNA expression, AR protein expression was investigated by IHC across all TNBC tumors from the Vanderbilt cohort (n=20). The percentage of tumor cells scored with nuclear AR staining and the intensity of staining were significantly higher in the LAR subtype (>10-fold; P<0.004) compared with all other TNBC subtypes (FIGS. 18A-18B). Hierarchical clustering was performed using an AR-activated gene signature (Chen C D, et al. Nat Med. 2004; 10(1):33-39) on the training and validation TNBC tumor data sets. Hierarchical clustering with this signature segregated the majority of LAR tumor profiles from other subtypes (FIGS. 19A-19B). Tumors in the LAR subtype display luminal GE patterns, with FOXA1, KRT18, and XBP1 among the most highly expressed genes (FIG. 20A). Others have previously described a breast cancer subgroup expressing AR termed molecular apocrine (Chen C D, et al. Nat Med. 2004; 10(1):33-39). While we do not have detailed pathology reports for all of the LAR tumors in this study, the TNBC subgroup GE centroids were used to correlate the apocrine samples from GSE-1561 (Farmer P, et al. Oncogene. 2005; 24(29):4660-4671; Table 1). The GE profiles of all 6 apocrine tumors (GSM26883, GSM26878, GSM26886, GSM26887, GSM26903, and GSM26910) described in the study strongly correlate with LAR, suggesting that the LAR TNBC subtype is composed of AR-driven tumors that include the molecular apocrine subtype.
Intrinsic molecular breast cancer subtype classification of TNBC. Breast cancers can be classified as luminal or basal-like, dependent on their expression of different cytokeratins. TNBC tumor subtypes display differential expression of both basal-like cytokeratins (KRT5, KRT6A, KRT6B, KRT14, KRT16, KRT17, KRT23, and KRT81) and luminal cytokeratins (KRT7, KRT8, KRT18, and KRT19) across the subtypes (FIGS. 20A-20B). The UNS, BL1, BL2, and M subtypes expressed higher levels of basal cytokeratin expression, while tumors within the LAR subtype lacked basal cytokeratin expression and expressed high levels of luminal cytokeratins and other luminal markers (FOXA1 and XBP1) (FIG. 20A). In addition to cytokeratin expression, breast cancers can be classified by an “intrinsic/UNC” 306-gene set into 5 major subtypes (basal-like, HER2-like, normal breast-like, luminal A, and luminal B) (Hu Z, et al. BMC Genomics. 2006; 7:96). Since TNBCs are largely considered basal-like, each of the 386 TNBC tumor profiles were correlated to the intrinsic gene set centroids of the 5 molecular subtypes, as previously described (Hu Z, et al. BMC Genomics. 2006; 7:96). Tumors were assigned to 1 of the molecular subtypes based on the highest correlation coefficient between each TNBC expression profile and the 5 molecular subtype centroids. This analysis resulted in 49% (n=188) of the TNBC training set classified as basal-like, 14% (n=54) as luminal A, 11% (n=42) as normal breast-like, 8% (n=31) as luminal B, 5% (n=19) as HER2, and 13% (n=52) as unclassifiable (FIG. 20B). This confirms that most TNBCs classify to the basal-like molecular subtype. Both the unstable tumors and BL1 tumors correlated strongly to the basal-like intrinsic molecular classification (76% and 85%, respectively). However, the BL2, IM, and M subtypes only moderately correlated to the basal-like molecular class (31%, 58%, and 47%, respectively), with a portion of tumors unclassified (22%, 17%, and 18%, respectively) (FIG. 20B). The M and MSL subtypes displayed the largest portion of tumors classified as normal breast-like (25%, and 46%, respectively). The BL2, M, and MSL subtypes were a mixture of classifications, suggesting the intrinsic classification system may not be suitable for characterizing these TNBC subtypes. The majority of TNBC tumors within the LAR subtype were classified as either luminal A or luminal B (82%), and none were classified as basal-like, further supporting the luminal origin of the LAR subtype (FIGS. 20A-20B). While only 49% of the tumors were classified as basal-like according to the intrinsic gene set, IHC staining performed on the Vanderbilt subset of tumors (n=25) showed that the majority (88%) of TNBCs stained positive for the basal cytokeratins CK5/6. Additionally, 56% of the Vanderbilt tumors stained positive for EGFR, similar to a previous study that found 56% of n=929 pooled from 34 studies that were positive for EGFR or CK5/6 (Yang X R, et al. J Natl Cancer Inst. 2011; 103(3):250-263). There were no statistical differences between CK5/6 and EGFR staining across TNBC subtypes. Thus, the majority of TNBCs display a basal-like phenotype by IHC, while only approximately half correlate to the basal-like intrinsic gene set.
Patient relapse-free survival and distant-metastasis-free survival differs among TNBC subtypes. Relapse-free survival (RFS) between TNBC subtypes was significantly different (log-rank test; P=0.0083) (FIG. 21A), despite variability of therapy and duration of treatment. RFS was significantly decreased in the LAR subtype compared with the BL1 (hazard ratio [Hr]=2.9), IM (Hr=3.2), and MSL (Hr=10.5) subtypes (P<0.05) (FIG. 21B). RFS was significantly decreased in the M subtype compared with BL1 (Hr=2.6) and IM (Hr=2.9), while the MSL subtype had higher RFS than the M subtype (FIG. 21B). Distant-metastasis-free survival (DMFS) did not vary between TNBC subtypes (log-rank test; P=0.2176), but the M subtype had a significantly higher Hr compared with the BL1 (Hr=2.4, P <0.05) and IM (Hr=1.9, P<0.06) subtypes (FIGS. 21C and 21D). The increased RFS in the absence of increased DMFS for patients in the LAR subtype suggests that recurrence is due to local relapse. Tumor size and grade were not significantly different among TNBC subtypes, but age at diagnosis was greater in the LAR subtype (P<0.0001), potentially contributing to decreased RFS of this subtype compared with other subtypes (FIGS. 22A-22B).
TNBC cell line models for TNBC subtypes. Cell line models would facilitate preclinical experiments to define differential drug sensitivity of the distinct subtypes within this heterogeneous disease. Using the bimodal filtering approach on ER, PR, and HER2 GE levels from 2 independent breast cancer cell line data sets (GSE-10890 and ETABM-157), 24 and 25 triple-negative cell lines were identified in the GSE-10890 and ETABM-157 GE data sets, respectively. Of the cell lines present in both data sets, nearly all had similar predictions of triple-negative status by bimodal filtering. Discrepancies in some cell lines (e.g., HCC1500 and HCC1937) may be the result of differences in culturing methods and/or loss of hormone receptor expression over time in culture. Analysis of these 2 data sets identified 30 nonoverlapping TNBC cell lines.
GE profiles from the cell lines were correlated to the centroids for each of the 6 TNBC subtypes. The majority of cell lines (27 of 30) were assigned to TNBC subtypes (Table 3), except for BT20, HCC1395, and SW527, which had low correlations (<0.1) or were similar between multiple subtypes (P >0.05). Of the 7 cell lines that contained known BRCA1 and BRCA2 mutations, 5 correlated with the BL1 or BL2 subtypes (HCC1937, HCC1599, HCC2157, HCC3153, and SUM149PT), consistent with this subtype containing tumors with defects in DNA repair pathways (Table 3) and gene ontologies enriched for GE involved in cell cycle and DNA repair functions (FIG. 13 ). Additionally, cell lines in the BL1 and BL2 subtypes were more genomically unstable, displaying significantly more chromosomal rearrangements than those in the mesenchymal subtypes (M and MSL) (average translocations, inversions, deletions=34 versus 14, respectively; P<0.01), as determined by SKY-FISH (www.path.cam.ac.uk/˜pawefish/index.html) (FIGS. 23A-23B). All cell lines that correlated to the basal-like intrinsic molecular subtype were in the BL1, BL2, or IM subtypes (Table 3). Only 2 cell lines (HCC1187 and DU4475) were placed into the IM subtype, suggesting that the IM subtype is underrepresented in cell culture.
Cell lines in the M and MSL subtypes were generated from highly dedifferentiated tumors derived from unique pathologies (e.g., HS578T, carcinosarcoma; and SUM159PT, anaplastic carcinoma) and expressed both epithelial and mesenchymal components. All cell lines assigned to the M and MSL subtype have spindle-like morphology in 2D culture (CAL-120, CAL-51, MDA-MB-157, MDA-MB-231, MDA-MB-436, SUM159PT, HS578T, and BT549) or stellate-like morphology in 3D (Kenny P A et al., Mol Oncol, 2007; 1:84-96). Five cell lines matched to the LAR subtype (MDA-MB-453, SUM185PE, HCC2185, CAL-148, and MFM-223).
Two distinct basal groups (A and B) have been identified by GE profiling of breast cancer cell lines (Neve R M, et al. Cancer Cell. 2006; 10(6):515-527). Basal A cell lines display epithelial characteristics and are associated with BRCA1 gene signatures, while basal B cell lines are more invasive and display mesenchymal and stem/progenitor-like characteristics. The GE analyses revealed that the majority of basal A cell lines belong to the BL1 and BL2 subtypes, while the majority of basal B cell lines fall into the M and MSL subtypes (Table 3). Hierarchical clustering analysis was tested on all TNBC cell lines using the most differentially expressed genes from the tumors to determine whether GE patterns of the cell lines are similar within TNBC subtypes (FIG. 24 ). Three clusters were identified: LAR, containing all 4 LAR lines; basal-like, containing lines in the BL1 and BL2 subtypes; and mesenchymal-like, containing lines in the M and MSL subtypes. This clustering analysis indicates that TNBC cell lines can be classified into 3 main groups: basal-like (BL1 and BL2); mesenchymal-like (M and MSL); and LAR. This classification will be used in subsequent sections.
TNBC cell lines have differential sensitivity to therapeutic agents. There are a variety of targeted therapies undergoing clinical investigation in patients with TNBC including those targeting PARP (Farmer H, et al. Nature. 2005; 434(7035):917-921), AR (Agoff N S et al. Am J Clin Pathol. 2003; 120(5):725-731), Src (Finn R S, et al. Breast Cancer Res Treat. 2007; 105(3):319-326), and PI3K/mTOR (Marty B, et al. Breast Cancer Res. 2008; 10(6):R101) signaling. The panel of TNBC cell lines was used to assess differential response to several agents targeting these pathways. For comparison primary human mammary epithelial cells (HMECs) were analyzed in cell viability assays. The half-maximal inhibitory concentration (IC50) values were determined for the following drugs [targets]: veliparib (ABT-888) [PARP1/2]; olaparib (AZD2281) [PARP1/2]; cisplatin [DNA]; bicalutamide [AR]; 17-DMAG [Hsp90]; dasatinib [Src/Abl]; and NVP-BEZ235 [PI3K and mTOR].
Cell-cycle and DNA damage response genes are elevated in the BL1 and BL2 subtypes (FIG. 13 ), and 5 of the 7 lines in the cell line panel that carry BRCA1/2-mutations reside in these subtypes (Table 3). Given the previous clinical trial observations with PARP inhibitors and cisplatin (Bhattacharyya A et al. J Biol Chem. 2000; 275(31):23899-23903; Jones P, et al. J Med Chem. 2009; 52(22):7170-7185; Evers B, et al. Clin Cancer Res. 2008; 14(12):3916-3925), it was predicted that these agents would preferentially decrease viability in cell lines with DNA repair defects that are representative of the BL1 and BL2 subtypes. The TNBC cell line panel was treated with the PARP inhibitors veliparib and olaparib, but not all cell lines representative of the basal-like TNBC subtypes were sensitive to PARP inhibition (FIGS. 4A-4D). The BRCA1-null cell line HCC1937 was sensitive to veliparib (IC50=4 μM) but not olaparib (IC50>100 μM), while the BRCA1-mutant MDA-MB-436 was sensitive to both PARP inhibitors (veliparib IC50=18 μM and olaparib IC50=14 μM). The BRCA2-mutant cell line HCC1599 lacked sensitivity to either PARP inhibitor (veliparib IC50>30 μM and olaparib IC50>100 μM). Thus, in addition to BRCA1/2 status, other properties of the tumor may dictate sensitivity to a given PARP inhibitor. Unlike PARP inhibitor sensitivity, basal-like lines were significantly more sensitive to cisplatin than mesenchymal-like lines (average IC50=81.1M vs. IC50=161.1M, P=0.032) or LAR lines (average IC50=81.1M vs. IC50=15 μM, P=0.017) (FIGS. 4 , E and F). The BRCA1-mutant cells (SUM149PT, HCC1937, and MDA-MB-436) and BRCA2-mutant cells (HCC1599) were among the most sensitive to cisplatin treatment (FIG. 4E).
Since cell lines representative of the LAR subtype (MDA-MB-453, SUM185PE, CAL-148, and MFM-223) express high levels of AR mRNA and protein (FIG. 25A), we compared the sensitivity of TNBC cell lines to the AR antagonist bicalutamide (FIGS. 5A and 5B). IC50 values for the majority of TNBC cell lines were not achieved using the highest dose of 500 μM. However, all LAR cell lines tested (SUM185PE, CAL-148, MDA-MB-453, and MFM-223) and a subset of mesenchymal-like cell lines that express low levels of AR (HS578T, BT549, CAL-51, and MDA-MB-231) were more sensitive to bicalutamide than basal-like cell lines (average IC50=227 μM vs. IC50>600 μM, P=0.007; and average IC50=361 μM vs. IC50>600 μM, P=0.038, respectively) (FIG. 5A).
Since AR requires the Hsp90 chaperone for proper protein folding and stability (Solit D B, et al. Clin Cancer Res. 2002; 8(5):986-993), the sensitivity of the cell line panel to an Hsp90 inhibitor, 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17-DMAG), was determined. Again, the LAR cell lines were more sensitive to 17-DMAG compared with the majority of basal-like (average IC50=16 nM vs. IC50=81 nM; P=0.004) and mesenchymal-like (average IC50=16 nM vs. IC50=117 nM; P=0.05) cell lines (FIGS. 5C and 5D), albeit 17-DMAG has many other targets. These data strongly suggest that LAR tumors are driven by AR signaling and AR represents a therapeutic target for this subtype. Importantly, AR status in TNBC patients represents a molecular marker for preselection of patients for antiandrogen therapy.
To determine whether the growth of the LAR cell lines was AR dependent, MFM-223, MDA-MB-453, and SUM185PE cell lines were treated with control and AR-targeting siRNA. Knockdown of AR in the experimental samples at the protein level was verified (FIG. 25B), colony growth assays were performed, and the number of colonies formed after 14 days of siRNA transfection were analyzed. The ability of all the LAR cell lines to form colonies was significantly reduced after knockdown of AR expression as compared with control samples (MFM-223=55%, P=0.031; MDA-MB-453=51%, P=0.004; and SUM185PE=42.3%, P=0.002) (FIG. 25C), indicating that AR expression is in part responsible for tumor cell viability/survival.
GE analysis of the mesenchymal-like subtypes demonstrated enrichment in the expression of genes that make up components and pathways associated with EMT (TGFβ, ECM-receptor interaction, ALK, Wnt/β-catenin, and Rac1) and those associated with cell motility (focal adhesion, integrin signaling, Rac1, striated muscle contraction, and regulation of actin by Rho GTPase) FIG. 3 ). Since the nonreceptor tyrosine kinase Src plays critical roles in cell migration and the mesenchymal-like subtypes are enriched in cell motility pathways, the effect of the Src inhibitor dasatinib on the panel of TNBC lines was analyzed. Cell lines belonging to the mesenchymal-like subtypes (M and MSL) were more sensitive to dasatinib than the LAR cell lines (average IC50=22 μM vs. IC50=88 μM, P=0.024) (FIGS. 6A and 6B).
Since activating mutations in PIK3CA are the most frequent genetic event in breast cancer (Samuels Y, et al. Science. 2004; 304(5670):554), the TNBC cell lines were treated with the dual PI3K/mTOR inhibitor NVP-BEZ235 (Maira S M, et al. Mol Cancer Ther. 2008; 7(7):1851-1863). TNBC cell lines that have activated PI3K/AKT signaling due to PIK3CA mutations or PTEN deficiency (Table 3) were highly sensitive to NVPBEZ235 (FIG. 6C). In addition, mesenchymal-like TNBC cell lines were more sensitive to NVP-BEZ235 compared with basal like cell lines (average IC50=44 nM vs. IC50=201 nM; P=0.001) (FIGS. 6C and 6D), which may suggest that deregulation of the PI3K pathway is important for this subtype. LAR cell lines were also more sensitive to NVP-BEZ235 compared with basal-like cell lines (average IC50=37 nM vs. 116 nM; P=0.01) (FIGS. 6C and 6D). This sensitivity can be explained by PIK3CA mutations, frequent in the LAR subtype, with all LAR cell lines containing PIK3CA-activating mutations (HCC2185, MDA-MB-453 CAL-148, MFM-223, and SUM185PE) (Table 3). While PIK3CA mutations predicted NVP-BEZ235 sensitivity, PTEN deficiencies (mutation or loss of protein expression) did not correlate with sensitivity.
Xenograft tumors derived from TNBC cell lines display differential sensitivity to therapeutic agents in vivo. In order to further analyze the susceptibility or resistance of TNBC subtypes to therapeutic agents in a more physiological setting than 2D culture, xenograft tumors were established in nude mice from cell lines representative of the basal like (HCC1806 and MDA-MB-468), mesenchymal-like (CAL-51 and SUM159PT), or LAR (SUM185PE and CAL-148) subtypes. After tumors reached an approximate volume of 25-50 mm3, the mice were treated with cisplatin, NVP-BEZ235 or bicalutamide. The sensitivity of the TNBC cell lines to the therapeutic agents when grown as 3D xenograft tumors in vivo was very similar to that seen with the cell lines grown in 2D monolayer culture. The xenograft tumors derived from the 2 cell lines representative of basal-like tumors (HCC1806 and MDA-MB-468) were highly and differentially sensitive to cisplatin and were significantly growth inhibited (P<0.0001) relative to treatment with vehicle control or the other experimental treatments (bicalutamide and NVP-BEZ235) (FIG. 7 ). The tumors derived from the LAR cell lines (SUM185PE and CAL-148) and the mesenchymal-like cell line that expresses low level AR protein (CAL-51) (FIGS. 25A-25C) displayed significant sensitivity to bicalutamide (FIG. 7 ). The xenograft tumors derived from all the cell lines carrying activating PIK3CA mutations (SUM185PE, CAL-148, CAL-51, and SUM159PT) had partial response or complete compared with NVP-BEZ235 (FIG. 7 ).
In summary, cell lines representative of the 6 TNBC subtypes display different sensitivities to a variety of agents, and importantly, these differences can be attributed to distinct expression of cellular components and presence of mutations in key oncogenes and tumor suppressors. The results have immediate clinical translation, as they provide a valuable platform and insights for ongoing and future clinical investigation. The data indicate that patients with basal-like TNBC should be treated with agents that engage DNA damage signaling response pathways; those with tumors expressing AR should receive bicalutamide alone or in combination with PI3K inhibitors; and those with mesenchymallike TNBC should be considered for trials exploring the activities of an Src antagonist in combination with a PI3K inhibitor.
Supplemental Results
K-means Clustering of a subset of five datasets that were identified as TNBC by IHC. Since TNBCs are not identified by GE in the clinical setting, a similar GE analysis was performed on a dataset from TNBCs identified using IHC (n=183) complied from five datasets: GSE7904 (n=22), GSE19615 (n=34), GSE20194 (n=67), E-TABM-158 (n=30) and GSE22513, GSE-XXX (n=30). This compilation included 136 tumors (74.3%) that were identified by our bimodal filtering analysis. K-means clustering performed on the most differentially expressed genes (SD>0.8) resulted in the identification of 5 TNBC subtypes (FIGS. 11A-11B). One subtype contained only three tumors and analysis of GE showed these tumors to have high levels of ER and PR FIG. 11B). These tumors were considered to be false negatives by IHC and were removed from further analysis. Comparison of these IHC-identified TNBC samples using genes differentially expressed from the six TNBC subtypes revealed similar patterns of gene enrichment (FIG. 12 ).
Discussion
Through GE analysis of 3247 breast cancers, demonstrate herein is that TNBCs can be reliably identified by filtering GE profiles for ER, PR, and HER2 mRNA levels. 587 TNBC GE profiles from 21 studies (training set=386 and validation set=201) were compiled. An 18% incidence rate of TNBC across the 21 independent studies (training and validation combined) was observed, similar to previously reported TNBC prevalence (Bertucci F, et al. Int J Cancer. 2008; 123(1):236-240; Rakha E A, et al. Clin Cancer Res. 2009; 15(7):2302-2310). k-means and consensus clustering of tumor profiles revealed that TNBC is composed of 6 stable subtypes enriched in distinct gene ontologies and GE patterns. Furthermore, using a GE signature derived from TNBC patient tumors, cell-line models for each of the TNBC subtypes were identified.
Previously, the majority (50%-90%) of TNBCs have been classified as basal-like either by IHC or by correlation to the intrinsic molecular breast cancer subtypes (Bertucci F, et al. Int J Cancer. 2008; 123(1):236-240; Rakha E A, et al. Clin Cancer Res. 2009; 15(7):2302-2310; Kreike B, et al. Breast Cancer Res. 2007; 9(5):R65). A previous TNBC study identified 5 distinct hierarchical clusters in which 91% (88 of 97) of TNBCs identified by IHC correlated to the basal-like subtype (Kreike B, et al. Breast Cancer Res. 2007; 9(5):R65). However, the study lacked molecular analysis of the tumors and conclusions were limited to clinical outcomes based on pathological markers. The relationship between TNBC and basal-like breast cancer remains controversial (Rakha E et al. Clin Cancer Res. 2008; 14(2):618). The proportion of TNBCs with basal-like GE in our study was 47%, resulting in a higher proportion of TNBCs that correlate with other molecular subtypes: luminal A (17%), normal breast-like (12%), luminal B (6%), HER2 (6%), or unclassified (12%). The study herein indicates that TNBC is not limited to tumors with a basal-like phenotype; rather it is a heterogeneous collection of tumors with distinct phenotypes, as evidenced by the diverse GE patterns and varying sensitivity of representative cell lines to the targeted therapies assessed in this study.
The BL1 and BL2 subtypes express high levels of genes involved in cell proliferation and DNA damage response, suggesting patients with basal-like tumors would benefit from agents that preferentially target highly proliferative tumors (e.g., anti-mitotic and DNA-damaging agents). Consistent with this notion, patients with basal-like tumors had up to a 4-fold higher pCR after taxane-based and radiation-based treatment as compared with patients with tumors that displayed characteristics of the ML or LAR subtypes (Bauer J A, et al. Clin Cancer Res. 2010; 16(2):681-690; Juul N, et al. Lancet Oncol. 2010; 11(4):358-365).
Nearly all of the cell lines with known mutations in BRCA1 and BRCA2 had GE patterns that correlated with the basal-like subtype, which is in agreement with the current view that BRCA-mutant tumors display a basal-like phenotype (Stefansson O A, et al. Breast Cancer Res. 2009; 11(4):R47). A number of non-BRCAmutant cell lines that correlated with the basal-like TNBC subtypes contained nearly 2-fold the number of chromosome rearrangements as all other subtypes. These findings suggest that a predominant characteristic of basal-like TNBC is genomic instability (Kwei K A et al. Mol Oncol. 2010; 4(3):255-266). Because of GE similarities between basal-like TNBC and BRCA1-mutation carriers, PARP inhibitors are currently being tested in clinical trials for TNBC (Silver D P, et al. J Clin Oncol. 2010; 28(7):1145-1153). Despite success in BRCA-null cells, there is not sufficient evidence for PARP inhibitor efficacy in BRCA1/2-mutant breast cancer cells (Farmer H, et al. Nature. 2005; 434(7035):917-921). PARP sensitivity across a panel of TNBC cell lines did not correlate with TNBC subtype alignment or BRCA1/2-status in this study using short-term cell viability assays (72 hours). However, the synthetic lethal effects of PARP inhibition in homologous recombination-deficient cells may require multiple cell divisions and may be more appropriately tested in long-term survival assays. Other studies show that a subset of BRCA1-mutant tumors lack large-scale genomic alterations and these tumors represent a distinct disease entity and may not be susceptible to PARP inhibition (Stefansson O A, et al. Breast Cancer Res. 2009; 11(4):R47). Previous analyses of PARP inhibitors in isogenic BRCA2+/+ and BRCA2−/− cell lines suggest that sensitivity to this targeted therapy is dependent on the molecular context of the DNA repair machinery and that DNA repair requiring homologous recombination involves multiple redundant pathways (Farmer H, et al. Nature. 2005; 434(7035):917-921; Fong P C, et al. N Engl J Med. 2009; 361(2):123-134). BRCA1 is a relatively large protein (1863 aa) that forms numerous complexes that may not be entirely disrupted when BRCA1 and BRCA2 are mutated, as opposed to BRCA-null cells. Despite an incomplete understanding of the molecular mechanism of the PARP inhibitors in vivo, these drugs have been proven to be highly effective in the clinical trial setting (41% objective response rate; 6.2 month progression-free survival) (Audeh M W, et al. J Clin Oncol. 2009; 27:5500).
It was also found that BRCA1-mutant and non-BRCA-mutant basal like cell lines had relatively higher sensitivity to cisplatin treatment compared with all other TNBC subtypes. The results are consistent with the observed 21% pCR in a clinical trial investigating neoadjuvant cisplatin as a single agent in a heterogeneous TNBC patient population (Garber J E, et al. Breast Cancer Res Treat. 2006; 105:S149; Telli M L, Ford J M. Clin Breast Cancer. 2010; 10 suppl 1:E16-E22). These data collectively suggest that the use of proliferation biomarkers such as Ki-67 and development of markers identifying defects in DNA damage response signaling could provide patient selection and tailored treatments for basal-like TNBC. Use of cisplatin as a “targeted” agent alone or in combination with antimitotics (taxanes) and/or radiation may benefit patients with this subtype and a current trial is underway with these agents (Mayer I A, et al. J Clin Oncol. 2010; 28(1):15).
The IM subtype is highly enriched in immune cell signaling. Other studies have described the presence of immune response gene signatures in ER-negative and medullary breast cancers (Bertucci F, et al. Cancer Res. 2006; 66(9):4636-4644; Teschendorff A E et al. Genome Biol. 2007; 8(8):R157). Similar to these studies, it was found elevated expression of T cell—associated genes, immune transcription factors, IFN regulatory factors, TNF, complement pathway, and antigen processing. It cannot be ruled out that the GE profile of the IM subtype comprises, at least in part, stromal components including immune cell infiltrate. However, the finding that the same proportion of microdissected tumors belongs to this group argues against stromal contamination.
The M and MSL subtypes share similar gene ontologies and GE profiles involving TGF-β, mTOR, Rac1/Rho, Wnt/β-catenin, FGFR, PDGFR, and VEGF signaling pathways. These signaling pathways are prominent in processes of EMT and stem cell-like properties of a CD44+CD24-population of normal mammary cells (Mani S A, et al. Cell. 2008; 133(4):704-715; Shipitsin M, et al. Cancer Cell. 2007; 11(3):259-273). Similarly, the MSL subtype is made up at least in part by the recently described claudin-low tumors, which lack luminal differentiation markers, high enrichment for EMT markers, immune response genes, and cancer stem cell-like features (Prat A, et al. Breast Cancer Res. 2010; 12(5):R68). Interestingly, the M and MSL subtypes differed clinically, with patients in the M subtype presenting with shorter RFS. This may be a reflection of differences in proliferation, as the M subtype displayed higher expression of proliferation-associated genes, including Ki-67. Additionally, patients with the M and MSL subtypes had decreased 5-year DMFS, consistent with enrichment in pathways associated with metastasis and motility.
Tumors within the mesenchymal-like subtypes have GE profiles that are similar to those from mesenchymal cells and metaplastic breast cancers (Hennessy B T, et al. Cancer Res. 2009; 69(10):4116-4124). Metaplastic breast cancers have lineage plasticity, including foci of spindle cell, as well as osseous or cartilaginous differentiation (Gibson G R, et al. Am Surg. 2005; 71(9):725-730). A recent study found that 47% of metaplastic breast cancers sequenced have PIK3CA mutations and have higher phosho-AKT expression (Hennessy B T, et al. Cancer Res. 2009; 69(10):4116-4124). It was found herein that TNBC mesenchymallike cell lines preferentially responded to the dual PI3K/mTOR inhibitor NVP-BEZ235. This response to NVP-BEZ235 was demonstrated in cell lines that carry PIK3CA mutations in xenografts in addition to an MSL cell line (SUM159PT) that lacks PIK3CA mutation or PTEN deficiencies, suggesting that the PI3K/mTOR pathway is important in the mesenchymal-like subtype.
The mesenchymal-like subtypes were enriched in pathways associated with EMT and cell motility. There is evidence of a prominent role for Src in tumor cells that are highly invasive, such as those that have undergone EMT (Guarino M. J Cell Physiol. 2010; 223(1):14-26). Accordingly, we found that mesenchymal-like TNBC cell lines had differential sensitivity to dasatinib. Markers of EMT may have clinical value for patient preselection for trials using dasatinib. In addition, Wnt-signaling pathways regulate EMT and may contribute to tumor cell invasion (Shin S Y, et al. Cancer Res. 2010; 70(17):6715-6724). Mutations in the Wnt/β-catenin pathway (CTNNB1, APC, and WISP3) occur frequently (52%) in metaplastic breast cancer, suggesting that deregulated Wnt/β-catenin pathway in these tumors may be a viable therapeutic target (Hayes M J et al. Clin Cancer Res. 2008; 14(13):4038-4044). Inhibitors of Wnt/β-catenin are of great interest and currently are in preclinical development (MacDonald B T, et al. Dev Cell. 2009; 17(1):9-26). Drugs targeting this pathway could be of value for treating mesenchymal-like TNBC.
The LAR subtype was readily subclassified by an AR gene signature and high levels of luminal cytokeratin expression. GE analysis of the LAR subtype is consistent with a prior report of a subset of ER-negative tumors expressing AR-regulated genes (Doane A S, et al. Oncogene. 2006; 25(28):3994-4008). In addition, Farmer et al. described an apocrine tumor subtype based on GE profiling that was characterized by AR expression distinguishing this tumor subtype from other basal-like tumors (Farmer P, et al. Oncogene. 2005; 24(29):4660-4671). In the GE analysis of tumors from 21 studies described herein, the prevalence of the LAR tumors was 11% (62 of 587) of TNBCs or 2% (62 of 3247) of all breast cancers. Analysis of clinical data demonstrated that patients in the LAR subtype had higher RFS but no difference in DMFS compared with all other TNBC subtypes, suggesting these patients have local relapse. The higher RFS could imply that this group of patients received ineffective therapies (standard chemotherapy); however, patients in the LAR group were significantly older at diagnosis and the extent of disease or age-associated comorbidities that affect the ability to deliver treatment as planned may have contributed to relapse. Older age at diagnosis has previously been reported in patients with AR-positive TNBC and is associated with postmenopausal status (Agoff N S et al. Am J Clin Pathol. 2003; 120(5):725-731). Whether this AR-driven subtype is arising from hormone-replacement therapy (HRT) merits further investigation; however, it is becoming clear that the risk of breast cancer increases with HRT, and synthetic progestins such as medroxyprogesterone acetate have been shown to bind and disrupt AR (Birrell S N et al. FASEB J. 2007; 21(10):2285-2293). As described herein, five (5) cell lines were identified that represent the LAR subtype and it was shown that they are sensitive to bicalutamide and 17-DMAG, suggesting that therapies targeting AR may be effective against tumors that express this hormone receptor. In fact, there is a clinical trial (NCT00468715) underway testing the effect of bicalutamide in preselected patients with ER/PR-negative AR-positive tumors. This further supports using in silico-based approaches to provide leads for trials with other targeted therapies in TNBC subtypes.
In addition to agents that target AR function, LAR cell lines were also sensitive to PI3K inhibition. This sensitivity correlated with PIK3CA mutations. All 5 LAR cells lines have activating PIK3CA mutations and are sensitive to the PI3K inhibitor NVP-BEZ235, similar to ER-positive breast cancer in which PIK3CA mutations are common (Gonzalez-Angulo A M, et al. Clin Cancer Res. 2009; 15(7):2472-2478; Stemke-Hale K, et al. Cancer Res. 2008; 68(15):6084-6091). These findings suggest simultaneous targeting of AR and the PI3K/mTOR pathway may be of clinical benefit for LAR TNBC patients, as this combination has been shown to be synergistic in AR-dependent prostate cancer cells (Liu X et al. J Clin Oncol. 2010; 28:(suppl; abstr e15049)).
The GE analysis of TNBC described herein demonstrates that with sufficient sample size, distinct subtypes of TNBC can be identified with putative molecular targets. The analyses provides biomarkers that can be used for patient selection in the design of clinical trials for TNBC as well as identification of potential markers of response to treatment. The identification of cell lines representing TNBC tumor subtypes provides key models for preclinical studies with newly developed targeted agents.
Example 2 Frequent PIKCA Mutations in AR-Positive TNBC Confer Sensitivity to PI3K and AR Inhibitors
Described herein is the further investigation of the molecular features of the LAR subtype to identify rationale combinations of drugs that would show robust efficacy against AR-positive TNBC cells with the added goal of generating preclinical data for rationale clinical trial design. It was discovered that PIK3CA kinase mutations were a frequent event in AR-positive TNBC tumors. It was found that genetic or pharmacological targeting of AR in LAR cells increased the therapeutic benefit of PI3K/mTOR inhibition. Further, it was discovered that after bicalutamide treatment of mice bearing LAR xenograft tumors, the PI3K pathway signaling increased. Thus, the combination of AR antagonism and PI3K/mTOR inhibition was examined and an additive or synergistic effect, depending on the TNBC cell line, was found. Given that AR expression is a robust biomarker for selection of LAR TNBC patients, the preclinical findings provide the rationale for preselecting AR-positive TNBC patients for treatment with AR inhibitors and/or P13K/mTOR inhibitors, and future clinical trials exploring the efficacy of combinations of drugs that target AR and PI3K signaling.
Results and Discussion
AR-positive TNBC tumors are enriched for PIK3CA kinase domain mutations. High frequency PIK3CA mutations in AR-positive TNBC cell lines are described in Example 1. To determine if PIK3CA mutation was the result of in vitro selection during establishment of the tumor-derived cell lines or if PIK3CA mutations were frequent in AR-positive TNBC tumors, performed Sanger sequencing was performed on 26 AR-positive TNBC cases. PCR-amplified regions from exons 9 and 20 that harbor the most frequently occurring activating mutations in PIK3CA were sequenced. Consistent with the observations in cell lines, nearly all of the detected mutations occurred at amino acid H1047 (20 of 21) with only one occurring at E545K. Mutation status was confirmed with digital droplet PCR on DNA amplified from exon 20. Using both Sanger sequencing of exons 9 and 20 and digital droplet PCR, PIK3CA mutations were significantly (P<0.0056) enriched in AR-positive TNBC tumors (17 of 26, 65.3%) versus AR-negative TNBC tumors (10 of 26, 38.4%).
Validation of PIK3CA Mutations in AR-Expressing TNBC
To verify that PIK3CA mutations were enriched in AR-expressing TNBC, RNA-seq, DNA-seq and reverse-phase protein array (RPPA) data available from The Cancer Genome Atlas (TCGA) breast cohort (Cancer Genome Atlas Network, Nature, 490:61-70 (2012)) was analyzed. Following removal of false negatives for ER, 66 TNBC samples were identified and classified according to molecular subtypes (Chen et al., Cancer informatics, 11:147-156 (2012)). The distribution of samples across the TNBC molecular subtypes was similar to that previously published (Lehmann et al., J Clin Invest, 121:2750-2767 (2011)). (BL1=14 (21.1%), BL2=6 (9.0%), IM=15 (22.7%), M=17 (25.7%), MSL=7 (10.6%) and LAR=7 (10.6%) (FIG. 26B). As expected, RNA-seq data confirmed significant levels of AR RNA (6.8 vs. 0.3, p<0.0001) and protein (1.7 vs. 0.35, p<0.0001) in the LAR subtype. Overall, PIK3CA mutations were relatively rare in TNBC cases in the TCGA dataset (4 of 66, 6.0%). However, when sorted by subtype, PIK3CA mutations were significantly enriched in LAR TNBC (3 of 7, 42.8%) compared to all other subtypes (1 of 59, 1.7%, p<0.0001), supporting our analysis of 26 TNBC cases above (FIG. 26B).
LAR TNBC Cell Lines have Increased PI3K Pathway Activation and are Sensitive to PI3K and PI3K/mTOR Inhibition.
RPPA was used to investigate AR, PTEN, active AKT (pS473 and PT308) and active GSK3β (pS21/pS9) protein levels in LAR cell lines to determine if the PI3K pathway was active in AR expressing, PI3K mutant TNBC. Activated PI3K signaling correlated with PIK3CA mutation and PTEN loss. AR-expressing TNBC cell lines expressed higher levels of active AKT as evidenced by increased pS473 (2.18 vs. −0.23, p=0.007) and pS308 (1.45 vs. −0.10, p=0.0032) and increased downstream phosphorylation of GSK3β at pS9 (1.00 vs. −0.26, p=0.0094) and pS21/pS9 (1.12 vs. −0.19, p=0.0026) (FIG. 27 ). Similarly, those cell lines that were null for PTEN (BT549, CAL51, HCC1395, HCC1937, HCC38, HCC70, MDA-MB-436 and MDA-MB-468) displayed higher levels of active PI3K signaling (p-AKT S473 1.28 vs. −1.28, p=0.0001, p-AKT T308 0.41 vs. −0.53, p=0.0018, p-GSK S9 0.37 vs. −0.76, p=0.0002 and pGSKαβ S9/S21 0.26 vs. −0.59, p=0.0009).
To validate the RPPA data, immunoblot analysis of the PI3K/mTOR pathway in four of the LAR cell lines, as well as primary cultures of normal mammary epithelial cells (HMEC) and the AR-dependent prostate cancer cell line LNCaP (FIG. 27 ), was also performed. AR receptor protein in the LAR TNBC cell lines was expressed at levels equal to or greater than LNCaP cells. AR-expressing cell lines displayed higher levels of phosphorylated AKT (S473) and phosphorylated ribosomal S6, confirming an activated PI3K pathway and downstream mTOR activation, respectively (FIG. 27 ).
To evaluate the clonality of AR expression, immunohistochemistry (IHC) for AR and p-AKT was performed on a cell line-based array (FIG. 27 ). The AR-expressing cell lines displayed differing percentage of cells positive for AR protein expression (CAL148: 20%, MDA-MB-453: 50%, MFM-223: 50% and SUM185: 90%). However, p-AKT expression was much more homogenous, indicating that activation of PI3K/AKT signaling was an earlier clonal event.
To determine the sensitivity of PIK3CA mutation-bearing LAR cell lines to PI3K inhibitors relative to other TNBC cell lines, a large panel of TNBC cell lines was treated with pan-PI3K inhibitors BKM120 (Novartis) and GDC-0941 (Genentech) or with the dual PI3K/mTOR inhibitors NVP-BEZ235 (Novartis) and GDC-0980 (Genentech). AR-expressing and other TNBC cell lines containing PIK3CA mutations were among the most sensitive to PI3K inhibition, as indicated by low half maximal inhibitory concentration (IC50) values (FIGS. 28A-28C-2 ). In contrast, PTEN deficiencies, while conferring PI3K pathway activation, did not predict for sensitivity to PI3K inhibitors (FIGS. 28A-28C-2 ). Nearly all of the PIK3CA mutant cell lines were more sensitive to PI3K/mTOR inhibitors than primary cultures of human mammary epithelial cells (HMEC).
Genetic and Pharmacologic Inhibition of AR Increases the Efficacy of PI3K Inhibition.
Whether the combined targeting of AR and PI3K would be more effective than either agent alone was investigated. To determine the effectiveness of combinatorial treatment both genetic knockdown and pharmacological inhibition of AR with shRNA and bicalutamide, respectively, in the presence and absence of PI3K inhibitors were performed (FIGS. 29A-29B). Immunoblot analysis confirmed decreased AR protein levels in three AR-expressing TNBC cell lines infected with lentiviral particles containing non-targeting shRNA or two AR-targeting hairpins (FIG. 29A). Both shRNA expression vectors targeting AR decreased cell viability across multiple doses of PI3K or PI3K/mTOR inhibitors (FIG. 29B). Efficacy of AR-targeting in combination with PI3K or PI3K/mTOR inhibitors was independent of compound, as similar results were obtained with BKM-120 and NVP-BEZ235 (FIG. 34 ).
Given that knockdown of AR increased the efficacy of PI3K inhibitors, pharmacological targeting of the AR was performed using bicalutamide. Both GDC-0941 and GDC-0980 displayed efficacy in AR-expressing TNBC cell lines alone and viability could be decreased further with the addition of 25 μM bicalutamide (CDX) across multiple concentrations of PI3K inhibitors (FIG. 30A). PI3K inhibition in combination with bicalutamide decreased cell viability in an additive or synergistic manner, as demonstrated by viabilities at, or below, the line of additivity. Immunoblot analysis was performed on cell lystates from untreated cells or cells treated alone or in combination with CDX, GDC-0941 and GDC-0980 to evaluate attenuation of the PI3K/mTOR pathway. Both GDC-0941 and GDC-0980 decreased activated AKT (p-5473), however, GDC-0980 was more effective at decreasing mTOR activity as measured by the decrease in levels of phosphorylated S6 (FIG. 30B). In contrast, bicalutamide (CDX) had minimal effect on either the PI3K/mTOR pathway or AR levels. Interestingly, GDC-0980 alone decreased AR levels and this decrease could be increased further by the addition of bicalutamide, suggesting that mTOR signaling contributes to the stability of AR protein, similar to crosstalk observed in prostate cancer (FIG. 30B) (Carver et al., Cancer Cell, 19:575-586 (2011)).
To determine if the decreased viability observed in AR-positive TNBC cell lines that were treated with the combination of pathway inhibitors described above was attributed to apoptotic cell death, activated caspase 3/7 levels after drug treatments were measured (FIGS. 31A-31B). Cells were treated with a high dose of adriamycin (ADR, 3□M) to measure the ability of cells to engage the apoptotic pathway. With exception of MFM-223 cells, each of the cell lines underwent robust caspase activation after 48 h of ADR treatment. While bicalutamide treatment did not induce caspase activation, treatment with both GDC-0941 and GDC-0980 alone caused caspase activation and the levels of apoptosis could be increased in combination with bicalutamide (FIG. 31A).
In addition to caspase activation, the percentage of sub 2N DNA (sub-G1)-containing cells (indicative of late stage apoptotic DNA fragmentation) was quantitated by FACs analysis. While bicalutamide treatment did not result in caspase activation, there was an increase in the sub-G1 fraction 48 h after treatment (5.25% to 18.44%) (FIG. 31B). Treatment with GDC-0941 or GDC-0980 resulted in higher sub-G1 fraction that increased in combination with bicalutamide, consistent with the caspase activation described above (FIG. 31B). Therefore, the decreased viability of AR-positive TNBC cell lines treated with a combination of AR and PI3K inhibitors can be, in part, attributed to apoptosis.
Simultaneous Targeting of AR and PI3K Decreases Viability of AR-Expressing Cell Lines Grown in a 3-D Forced Suspension Assay.
Since cell viability and drug sensitivity can be influenced by 2-D versus 3-D growth conditions, combined targeting of AR and PI3K/mTOR was evaluated in a 3-D forced suspension assay devoid of matrix. Cell lines were grown in forced suspension in agarose coated 96-well plates and treated with GDC-0941 or GDC0980 alone or in combination with bicalutamide (FIGS. 32A-32C). Increasing doses of GDC-0941 and GDC-0980 alone decreased cell aggregate size and viability in AR-expressing TNBC cell lines to a lesser degree when compared to similar treatments in 2-D (FIGS. 32A and 32B). However, the addition of bicalutamide was either additive or synergistic with PI3K/mTOR inhibition (FIG. 32B). Similar results were obtained when PI3K was inhibited with BKM120 or PI3K and mTOR were targeted with NVP-BEZ235 (FIG. 35 ). Together, these results indicate that simultaneous targeting of AR and PI3K/mTOR is more effective than either agent alone in AR-expressing TNBC.
PI3K is Activated after Pharmacological Inhibition of AR.
As described in Example 1, evaluation of bicalutamide in AR-expressing TNBC xenografts demonstrated significant reduction in tumor burden, however over time tumor growth occurred in the presence of drug, consistent with evolution of bicalutamide-resistant tumor cells. Since crosstalk between AR and PI3K has been demonstrated in prostate cancer, whether the level of PI3K activity changed between untreated and bicalutamide-resistant xenograft tumor cells was examined. Phospho-AKT (S473) levels were elevated in bicalutamide-resistant xenograft tumor cells (FIG. 37 ). Consistent with these findings were the findings herein of dose-dependent increases in p-AKT in cell lines upon genetic silencing of AR by shRNA (FIG. 38B).
Simultaneous Inhibition of AR and PI3K Signaling Decreases the Growth of LAR Cell Line-Derived Xenograft Tumors.
In order to further validate and investigate the combined targeting of AR and PI3K in AR-expressing TNBC in vivo, xenograft tumors in nude mice was established using two representative AR-positive TNBC cell lines (CAL-148 and MDA-MB-453). Following tumor establishment (25-50 mm3), mice were either treated with vehicle, bicalutamide, NVP-BEZ235, GDC-0980 alone or either NVP-BEZ235 or GDC-0980 in combination with bicalutamide. While each single agent alone significantly decreased tumor growth, both PI3K/mTOR inhibitors in combination with bicalutamide inhibited tumor growth to the greatest extent (FIG. 32C). Thus, the combined inhibition of AR and PI3K appears to be a rational treatment strategy for AR-expressing TNBC and should be considered in future clinical trial design.
Discussion
Herein we show that activating mutations in PIK3CA are a frequent event in a subset of AR-expressing TNBC (LAR) that display luminal gene expression patterns. The data herein provide the preclinical rationale for targeting AR in combination with PI3K/mTOR inhibition.
Higher levels of p-AKT in cell line xenografts after prolonged treatment with bicalutamide and increased p-AKT after siRNA knockdown of AR in cell lines is demonstrated herein. The data show that combined targeting of AR and PI3K pathway inhibition can overcome the PI3K pathway activation after AR inhibition. These studies demonstrate that both pathways cross-regulate each other by reciprocal feedback and coordinately support survival when either is repressed. The feedback between the AR and PI3K pathway may, in part, explain the enrichment of PIK3CA mutations observed in AR-positive TNBC, as this co-evolution may be necessary to overcome the increased PTEN levels driven by AR signaling.
Methods
PIK3CA Mutation Evaluation.
Two independent methods were used to identify PIK3CA mutations in tumor samples. Samples were identified as mutant if they had greater than 5% of the cells positive by microdroplet PCR, as there was complete concordance with Sanger sequencing calls, or called mutant by Sanger when microdroplet samples were 5% and >1% (FIGS. 33A-33B).
Sanger
To detect mutations in PIK3CA (H1047), a 588 bp fragment was PCR-amplified from exon 20 of PIK3CA using the following amplification primers; foreword: TGACATTTGAGCAAAGACCTG (SEQ ID NO: 1), reverse: CATAACATGAAATTGCGCATT (SEQ ID NO: 2). Sanger sequencing was performed on the PCR amplicon using the following internal sequencing primers; Exon Forward: ACATCATTTGCTCCAAACTGA (SEQ ID NO: 3) Reverse: CCTATGCAATCGGTCTTTGC (SEQ ID NO: 4) (Samuels et al., Science, 304:554 (2004)).
Droplet Digital PCR
The TaqMan PCR reaction mixture was assembled from a 2×ddPCR Mastermix (Bio-Rad), 20× primer, and probes (final concentrations of 900 and 250 nM, respectively) and template (variable volume) in a final volume of 20 μL. Each assembled ddPCR reaction mixture was then loaded into the sample well of an eight-channel disposable droplet generator cartridge (Bio-Rad). A volume of 70 μL of droplet generation oil (Bio-Rad) was loaded into the oil well for each channel. The cartridge was placed into the droplet generator (Bio-Rad). The cartridge was removed from the droplet generator, where the droplets that collected in the droplet well were then manually transferred with a multichannel pipet to a 96-well PCR plate. The plate was heat-sealed with a foil seal and then placed on a conventional thermal cycler and amplified to the end-point (40 cycles). Thermal cycling conditions were 95° C.×10 min (1 cycle), 94° C.×30 s and 57° C.×60 s (40 cycles), and 12° C. hold. After PCR, the 96-well PCR plate was loaded on the droplet reader (Bio-Rad), which automatically reads the droplets from each well of the plate. Analysis of the ddPCR data was performed with QuantaSoft analysis software (Bio-Rad) that accompanied the droplet reader. Primer sequences: Forward: 5′GATAAAACTGAGCAAGAGGCTTTGG′ (SEQ ID NO: 5), Reverse: 5′GCTGTTTAATTGTGTGGAAGATCCAA′ (SEQ ID NO: 6), Wild type probe: 5′-VIC-CCACCATGATGTGCA-MGB-3 (SEQ ID NO: 7), ‘H1047L probe:5′-FAM-CCACCATGATGAGCA-MGB-3′ (SEQ ID NO: 8), H1047R probe: 5′-FAM-CCACCATGATGCGCA-MGB-3′ (SEQ ID NO: 9).
Subtyping TNBC Cases and Validating PIK3CA Mutations in the TCGA.
RNA-seq (lvl 3), somatic mutations (lvl2) and reverse phase protein array (RPPA, lvl3) data for breast cancer was downloaded from The Cancer Genome Atlas (tcga-data.nci.nih.gov/). RNA-seq data (RPKM) was combined and molecularly subtyped using the online TNBCtype software according to published methods (Chen et al., Cancer Informatics, 11:147-1556 (2012)). After removal of potential ER positive, 137 TNBC samples were identified and assigned a molecular subtype.
Reverse Phase Protein Array on TNBC Cell Lines.
Tumor or cell lysates were two-fold-serial diluted for 5 dilutions (from undiluted to 1:16 dilution) and arrayed on nitrocellulose-coated slide in 11×11 format. Samples were probed with antibodies by CSA amplification approach and visualized by DAB colorimetric reaction. Slides were scanned on a flatbed scanner to produce 16-bit tiff image and the density was quantified by MicroVigene. Relative protein levels for each sample were determined by interpolation of each dilution curves from the standard curve (supercurve) of the slide (antibody) as previously described (Zhang et al., Bioinformatics, 25:650-654 (2009)). These values were Log 2 transformed and normalized for protein loading by transformation a to linear value. Linear normalized data was then median-centered for heatmap comparisons.
Cell Proliferation/Viability Assays and IC50 Determinations.
All cell lines were maintained in culture as previously described (JCI ref). Breast cancer cell lines and HMECs were seeded (3,000-10,000 cells) in quadruplicate wells in 96-well plates. Media was then replaced with either fresh media (control) or media containing half-log serial dilutions of the following drugs: GDC-0941 (10-nM-3000 nM), GDC0980 (1 nM-300 nM), BKM120 (10-nM-3000 nM) and NVP-BEZ235 (1 nM-300 nM) purchased from Selleck Chemicals (Houston, TX) or in combination with bicalutamide (Sigma, St. Louis, MO). Viability was determined from measuring fluorescent intensity after metabolic reduction of Alamar Blue in the presence/absence of drug after 72 hours. Viability assays were performed in triplicate and replicates were normalized to untreated wells. Inhibitory concentration (IC50) values were determined after double-log transformation of dose response curves as previously described (Lehmann et al., J Clin Invest, 121:2750-2767 (2011)).
Quantification of Apoptosis
Caspase 3/7 Activity
Cell lines were seeded in triplicate in 96-well plates. Media was removed and replaced with media containing vehicle (control) or indicated drugs. After 48 hrs, apoptosis was determined by measuring luciferase from activated caspase 3/7 upon addition of Caspase-Glo reagent (Promega, Madison, WI). Relative levels of caspase activity were normalized to viable cell number determined by metabolic reduction of Alamar blue.
shRNA Knockdown of AR
293FT cells were transfected with Lipofectamine 2000 (Invitrogen, Grand Island, NY) the packaging vectors PAX2 and pMD2.g (Addgene, Cambridge, MA) along with pLKO.1-puro Misson shRNA constructs (Sigma) targeting AR or a non-targeting control. Viral media was harvested 48 h post-transfection and added to target cells along with polybrene (10 μg/mL). 72 h post-infection MDA-MB-453 (8,000 cells/well), CAL-148 (5,000 cells/well) and MFM-223 (7,000 cells/well) seeded in quadruplicate in 96-well plates. Following overnight attachment media was replaced with fresh media (control) or media containing half-log dilutions of drugs. Following incubation in drug for 72 h viability was measured with alamarBlue as previously described (Lehmann et al., J Clin Invest, 121:2750-2767 (2011)).
Forced Suspension Viability Assay
CAL-148, MFM-223 and MDA-MB-453 cells were plated (10,000 cells/well) in quadruplicate into 96-well plates coated with 0.9% agarose diluted in corresponding media. Following 5 d of growth, cells were then treated with increasing half-log concentrations of GDC-0941 (10-3000 nM), GDC0980 (1 nM −300 nM), BKM120 (10 nM-3000 nM) and NVP-BEZ235 (1 nM-300 nM) alone or in combination with 25 μM bicalutamide (CDX) for an additional 72 h, upon which cells were imaged and viability determined by Alamar blue.
Immunostaining
Formalin fixed paraffin embedded (FFPE) tissue were used for immunohistochemical studies. Androgen receptor (AR) expression and p-AKT expression were evaluated in FFPE tissue using the DAKO (Carpinteria, CA) antibody: AR (clone AR411) at a 1:50 dilution and AKT p473 (Cell signaling, CA) at a 1:200 dilution for 1 h at room temperature. FFPE tissue was subject to antigen retrieval with high pH buffer (pH 8.0) followed by overnight incubation with an AR (1:30) or Ki67 (1:75) antibody dilution overnight.
Immunoblotting
Cells were trypsinized, lysed and relative protein expression was determined by Western blot as previously described (JCI reference) with the following antibodies; the AR polyclonal antibody, SC-N20 (Santa Cruz Biotechnology), pS6-Ser235/236 (Cell Signaling #4856), S6 (Cell Signaling #2317), pAKT-5473 (Cell Signaling #9271), AKT (Cell Signaling #9272), PARP (Cell Signaling #9542) and GAPDH (Millipore).
Xenograft Tumor Studies.
Five-week-old female athymic nude-Foxnlnu mice (Harlan Sprague-Dawley) were injected (s.c.) with either ˜4×106 (CAL-148) or ˜10×106 (MDA-MB-453) cells suspended in media (200 μL) into each flank using a 22-gauge needle. Once tumors reached a volume of 25-50 mm3, mice were randomly allocated to treatment or vehicle arms. Treatments included bicalutamide p.o. (100 mg/kg/d), GDC0980 p.o. (7.5 mg/kg/d) in 5% DMSO, 0.5% carboxymethyl cellulose (MCT) or NVP-BEZ235 p.o. (50 mg/kg/d) in 0.5% MCT or the combination of bicalutamide and either GDC-0980 or NVP-BEZ235 according to the above concentrations. Tumor diameters were serially measured at the indicated times with digital calipers and tumor volumes were calculated as width2×length/2.
Supplemental Results
Validation of Digital Droplet PIK3CA Mutant PCR Primers
Primers were validated using on cell lines with known PIK3CA status cells. The percentage of DNA reflected the frequency of mutant PIK3CA alleles similar to expected genotype status; HMECwt/wt=0.01%, MDA-MB-453 wt/mut=61.19% and SUM-185mut/mut=97.09% (FIG. 26A). The slightly higher mutant frequency in the heterozygous mutant cell line MDA-MB-453 is likely caused by chromosomal aneuploidy. Digital droplet PCR was far more quantitative than Sanger sequencing and both methods were 100% concordant when mutant DNA was present at levels greater than 5% (FIGS. 33A-33B).
Example 3
Cell proliferation/viability assays and IC50 determinations. All cell lines were maintained in culture as previously described herein. Breast cancer cell lines and HMECs were seeded (3,000-10,000 cells) in quadruplicate wells in 96-well plates. Media was then replaced with either fresh media (control) or media containing half-log serial dilutions of the following drugs: Sorafenib, OSi-906, BMS-754807 and PF2341066 purchased from Selleck Chemicals (Houston, TX) Viability was determined from measuring fluorescent intensity after metabolic reduction of Alamar Blue in the presence/absence of drug after 72 hours. Viability assays were performed in triplicate and replicates were normalized to untreated wells. Inhibitory concentration (IC50) values were determined after double-log transformation of dose response curves.
PDGFR inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines. Bar graph displays the half-maximal inhibitory concentration (nM) of the PDGFR inhibitor sorafenib for a panel of TNBC cell lines. Colorbar below designates the TNBC subtype (basal-like=black, mesenchymal-like=grey) or normal cells (white).
IGF1R inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines. Bar graph displays the half-maximal inhibitory concentration (nM) of the IGF1R inhibitor OSI-906 for a panel of TNBC cell lines. Colorbar below designates the TNBC subtype (basal-like=black, mesenchymal-like=grey) or normal cells (white).
IGF1R inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines. Bar graph displays the half-maximal inhibitory concentration (nM) of the IGF1R inhibitor BMS-754807 for a panel of TNBC cell lines. Colorbar below designates the TNBC subtype (basal-like=black, mesenchymal-like=grey) or normal cells (white).
MET inhibition selectively inhibits viability of mesenchymal-like TNBC cell lines. Bar graph displays the half-maximal inhibitory concentration (nM) of the MET inhibitor PF2341066 for a panel of TNBC cell lines. Colorbar below designates the TNBC subtype (basal-like=black, mesenchymal-like=grey) or normal cells (white).
TABLE 1
Breast cancer GE data sets used to derive the TNBC training set
False Positive (%)
Data Set Country Case Definitions BC cases IHC N (% TN) ER PR HER Refs.
GSE-494A Sweden Breast cancers from patients treated in Uppsala County, 251 ER, PR  25(10) 3.6 2.8 Miller LD, et al. An expression signature for
Sweden, between 1987 and 1989 p53 status in human breast cancer predicts
mutation status, transcriptional effects, and
patient survival. Proc Natl Acad Sci U.S.A.
2005; 102(38): 13550-13555.
GSE-904A USA Frozen tissue samples from primaiy, sporadic and 4 43 ER, PR,  17(40) 0.0 0.0 0.0 Richardson AL, et al. X chromosomal
BRCA1-mutant breast obtained through the Harvard HER2 abnormalities in basal-like human breast
Breast SPORE cancer. Cancer Cell. 2006; 9(2): 121-132.
GSE-2109A,B USA The international genomics consortium collection of 351 Variable0  60(17)
breast cancer tissue samples processed by the expO
biospecimen repository, obtained from community
hospitals
GSE-7390A,D Europe Lymp node-negative breast cancer patients, systemically 198 ER  29(15) 2.5 Desmedt C, et al. Strong time dependence of
untreated, performed at the Bordet Institute and part the 76-gene prognostic signature for node-
of the TRANSBIG consortium negative breast cancer patients in the
TRANSBIG multicenter independent validation
series. Clin Cancer Res. 2007; 13(11): 3207-3214.
E-TABM-15gE USA Frozen tumors collected from patients that received 100 ER, PR,  30(23) 0.8 2.3 2.3 Chin K, et al. Genomic and transcriptional
adjuvant chemotherapy (typically Adriamycin and CTX) HER2 aberrations linked to breast cancer
at UC San Francisco and the California Pacific Medical pathophysiologies. Cancer Cell.
Center between 1989 and 1997 2006; 10(6): 529-541.
Gse-2034A Netherlands Lymph node-negative relapse-free patients (80) and lymph 286 ER  53(19) 3.1 Wang Y, et al. Gene-expression profiles to
node-negative patients that developed a distant metastasis predict distant metastasis of lymph-node-
(106) treated at Erasmus Medical Center negative priinaiy breast cancer. Lancer.
2005; 365(9460): 671-679.
GSE-2990A Sweden, Invasive breast cancer from Uppsala, Sweden (88), of 189 ER  11(6) 1.1 Sotiriou C, et al. Gene expression profiling in
United which 64 received tamoxitan only, or from John breast cancer: understanding the molecular
Kingdom Radcliffe, Oxford, United Kingdom (101) of which all basis of histologic grade to improve prognosis.
received mixed hormone and chemotherapy J Natl Cancer Inst. 2006; 98(4): 262-272.
GSE-1456A Sweden Breast cancer patients receiving surgery at Kanolinska 159 NA  22(14) Pawitan Y, et al. Gene expression profiling
Hospital between 1994 and 1996 spares early breast cancer patients from
adjuvant therapy: derived and validated in two
population-based cohorts. Breast Cancer Res.
2005; 7(6): R953-R964.
GSE-22513A, USA Invasive breast cancer pretreatrnent biopsies taken from 112 ER, PR,  25(22) 0.0 0.0 0.0
GSE-28821, patients at the Vanderbilt-Ingram Cancer Center HER2
GSE-28796
GSE-11121A Germany Lymph node-negative breast cancer patients treated at 200 NA  19(10) Schmidt M, et al. The humoral immune system
the Johannes Gutenberg University Mainz between 1988 has a key prognostic impact in node-negative
and 1998. Patients were all treated with surgery without breast cancer. Cancer Res. 2008; 68(13): 5405-
adjuvant therapy 5413.
GSE-2603A USA Primary breast cancers surgically resected at the 99 ER, PR  32(32) 0.0 0.0 Minn AJ, et al. Genes that mediate breast
Memorial Sloan-Kcttcring Cancer Center cancer metastasis to lung. Nature.
2005; 436(7050): 518-524.
MDA133F USA Tumors from prcopcrative paclitaxel and 5-FU, 133 ER, PR,  21(16) 0.0 3.0 0.8 Gibson GR, Qian D, Ku JK, Lai LL.
doxonibicin. and CTX performed at the University HER2 Metaplastic breast cancer: clinical features and
of Texas MD Anderson Cancer Center outcomes. Am Surg. 2005;71(9): 725-730.
GSE-5364A Singapore Tumors from the National Cancer Centre of S 183 NA  31(17) Yu K. et al. A precisely regulated gene
ingapore expression cassette potently modulates
metastasis and survival in multiple solid
cancers. PLoS Genet. 2008;4(7): el000129.
GSE-1561A Belgium Tumors from patients with invasive or inflammatory 49 ER, PR  16(33) 0.0 0.0 Farmer P, et al. Identification of molecular
breast cancers treated with 5-FU, epirubicin, and CTX apocrine breast tumours by microarrav analysis.
obtained by the European Organization for Research and Oncogene. 2005;24(29): 4660-4671.
Treatment, Brussels, Belgium
TOTAL
2353 386(16) 1.7 1.7 .09
TABLE 2
Breast cancer GE data sets used to derive the TNBC validation set
False Positive (%)
Data Set Country Case Definitions BC cases IHC N (% TN) ER PR HER Refs.
GSE-5327A,B Netherlands Tumors from patients with lymph node-negative 58 N/A 17(30) Minn AJ, ct al. Genes that mediate breast
breast cancer, who did not receive systemic cancer metastasis to lung. Nature.
neoadjuvant or adjuvant therapy (EMC-344) at 2005; 436(7050): 518-524.
Erasmus Medical Center, Rotterdam, Netherlands
treated between 1980 and 1995
GSE-5874A USA Primary breast cancers between 1993 and 2003 95 ER, HER2 17(35) 8.3 0.0 Boersma BJ, ct al. A stromal gene signature
from patients receiv ing neoadjuvant associated with inflammatoiy breast cancer.
chemotherapy collected at Baltimore hospitals Int J Cancer. 2008; 122(6): 1324-1332.
GSE-12276A Netherlands Lymph node-negative breast cancers from patients 204 N/A 46(24) Bos PD, et al. Genes that mediate breast
with metastatic disease (114) following relapse cancer metastasis to the brain. Nature.
after chemotherapy (EMC-192) and pateints (48) 2009; 459(7249): 1005-1009.
without adjuvant systemic therapy (EMC-286) at
Erasmus Medical Center, Rotterdam, Netherlands
GSE-16446A,D Europe Tumors from patients with ER-negative primary 120 HER 46(38) 2.5 Li Y, et al. Amplification of LAPTM4B
breast cancers treated with single-agent and YWHAZ contributes to chemotherapy
neoadjuvant cpirubicin at European hospitals and resistance and recurrence of breast cancer.
coordinated at the Institut Jules Borde, Brussels, Nat Med. 2010; 16(2): 214-218.
Belgium
GSE-18864A,C USA Tumors from patients with stage II or III TNBCs 24 ER, PR, 24(100) Li Y, et al. Amplification of LAPTM4B
treated with cisplatin at the Dana-Farber/Harvard HER2 and YWHAZ contributes to chemotherapy
Cancer resistance and recurrence of breast cancer.
Nat Med. 2010; 16(2): 214-218.
GSE-19615A USA Breast cancers from patients and treated with 115 ER, PR, 30(26) 0.0 0.9 3.5 Li Y, et al. Amplification of LAPTM4B
adjuvant chemotherapy obtained from the HER2 and YWHAZ contributes to chemotherapy
Harvard Breast SPORE, Boston, resistance and recurrence of breast cancer.
Massachusetts, diagnosed between 2000 and Nat Med. 2010; 16(2): 214-218.
2003
GSE-20194A USA Pretreatment tumors obtained from stage I-III 278 ER, PR, 27(23) 2.6 4.3 0.0 Juul N, et al. Assessment of an RNA
breast cancers at the University of Texas HER2 interference screen-derived mitotic and
M.D. Anderson Cancer Center ceramide pathway metagene as a predictor
of response to neoadjuvant paclitaxel for
primary triple-negative breast cancer: a
retrospective analysis of five clinical trials.
Lancet Oncol. 2010; 11 (4): 358-365.
TOTAL 894 N/A 201(22) 1.7 2.6 1.7
ASource: GEO.
BGSE-5327 and GSE-16446 data sets are derived from ER-negative tumors by IHC.
CGSE18864 data set includes only ER, PR and HER2 negative tuomrs by IHC.
TABLE 3
Assignment of TNBC cell lines to subtypes
TNBC Subtype Correlation Intrinsic Basal
Subtype Cell Line (P Value) Histology MutationsA SubtypeB SubtypeC
Basal-like
BL1 HCC2157 0.66 (0.00) DC BRCA1; STAT4; UTX Basal Basal A
HCC1599 0.62 (0.00) DC BRCA2: TP53; CTNND1; TOP2B; Basal Basal A
CAMK1G
HCC1937 0.28 (0.00) DC BRCA1; TP53; MAPK13; MDC1 HER2 Basal A
HCC1143 0.26 (0.00) IDC TP53 Basal Basal A
HCC3153 0.24 (0.00) BRCA1 Basal A
MDA-MB-468 0.19 (0.06) DC PTEN, RB1; SMAD4; TP53 Basal A
HCC38 0.19 (0.01) DC CDKN2A; TP53 Unclassified Basal B
BLs SUM149PT 0.30 (0.00) INF BRCA1 Unclassified Basal B
CAL-851 0.25 (0.00) IGA RB1; TP53 Basal
HCC70 0.24 (0.04) DC PTEN; TP53 Basal Basal A
HCC1806 0.22 (0.00) ASCC CDKN2A; TP53; UTX Unclassified Basal A
HDQ-P1 0.18 (0.17) IDC TP53
HCC1187 0.22 (0.00) DC TP53; CTNNA1; DDX18; HUWE1; Basal Basal A
NFKBIA
DU4475 0.17 (0.00) DC APC; BRAPL MAP2K4; RB1 Unclassified
Mesenchymal-like
M BT-549 0.21 (0.00) IDC PTEN; RB1; TP53 Unclassified Basal B
CAL-51 0.17 (0.00) AC PIK3CA Luminal B
CAL-120 0.09 (0.00) AC TP53 Unclassified Basal B
MSL HS578T 0.29 (0.00) CS CDKN2A; HRAS; TP53 Unclassified Basal B
MDA-MB-157 0.25 (0.00) MBC NF1; TP53 Unclassified Basal B
SUM159PT 0.14 (0.00) ANC PIK3CA; TP53; HRAS Unclassified Basal B
MDA-MB-436 0.13 (0.00) IDC BRCA1; TP53 Unclassified Basal B
MDA-MB-231 0.12 (0.00) IDC BRAP; CDKN2A; KRAS; NF2; Unclassified Basal B
TP53; PDGFRA
LAR
LAR MDA-MB-453 0.53 (0.00) AC PIK3CA; CDH1; PTEN Luminal A Luminal
SUM185PE 0.39 (0.00) DC PIK3CA Luminal A Luminal
HCC2185 0.34 (0.00) PIK3CA Luminal A Luminal
CAL-148 0.32 (0.00) AC PIK3CA; RB1; TP53; PTEN Luminal A
MFM-223 0.31 (0.00) AC PIK3CA; TPS 3 Luminal A/B
Unclassified HCC1395 DC ATR; BRCA2; CDKN3A PTEN: Basal
FGFR1; PDGFRB; TP53
BT20 IDC CDKN2A; PIK3CA; TP53 HER2 Basal A
SW527 Luminal B
ASource: mutations taken from COSMIC database (www.sanger.ac.uk/genetics/CGP/cosmic/).
BMolecular subtype determined by correlation with UNC/intrinsic breast centroids (29).
CBasal subtype obtained from Neve RM et al. (32).
AC, adenocarcinoma;
ANC, anaplastic carcinoma;
ASCC, acantholytic squamous cell carcinoma;
C, carcinoma;
CS, carcinosarcoma;
DC, ductal carcinoma;
IDC, invasive ductal carcinoma;
IGA, invasive galactopharic adenocarcinoma;
INF, inflammatory ductal carcinoma;
MC, metaplastic carcinoma and MBC, medullaiy breast cancer.
TABLE 4
Top gene ontologies for TNBC subtypes in the training and validation datasets
Subtype Training Set (386) Validation Set (201)
BL1 DNA REPLICATION REACTOME CELL CYCLE KEGG
CELL CYCLE KEGG DNA REPLICATION REACTOME
RNA POLYMERASE G2 PATHWAY
CELL CYCLE KEGG METHIONINE METABOLISM
GLYCOSPHINGOLIPID BIOSYNTHESIS NEO CHOLESTEROL BIOSYNTHESIS
LACTOSERIES CELL CYCLE
ATR EBRCA PATHWAY RNA POLYMERASE
UBIQUITIN MEDIATED PROTEOLYSIS ATREBRCA PATHWAY
G2 PATHWAY G1 TO S CELL CYCLE REACTOME
PROTEASOME CASPASE CASCADE
G1 TO S CELL CYCLE REACTOME GLUTAMATE METABOLISM
METHIONINE METABOLISM AMINOACYL TRNA BIOSYNTHESIS
FAS PATHWAY GLYCOSPHINGOLIPID BIOSYNTHESIS NEO LACTOSERIES
CHOLESTEROL BIOSYNTHESIS G1 AND S PHASES
PYRIMIDINE METABOLISM CYSTEINE METABOLISM
AMINOACYL TRNA BIOSYNTHESIS ATM PATHWAY
DNA POLYMERASE MRNA PROCESSING REACTOME
CARBON FIXATION BIOSYNTHESIS OF STEROIDS
ALANINE AND ASPARTATE METABOLISM KREBS TCA CYCLE
PROTEASOME PYRIMIDINE METABOLISM
CELLCYCLE PATHWAY PROTEASOME PATHWAY
CDMAC PATHWAY SELENOAMINO ACID METABOLISM
KREBS TCA CYCLE CELL CYCLE PATHWAY
ALANINE AND ASPARTATE METABOLISM
GAQ PATHWAY
AMINOACYL TRNA BIOSYNTHESIS
FAS PATHWAY
PROTEASOME
BL2 PORPHYRN AND CHLOROPHYLL METABOLISM BLADDER CANCER
GATA3 PATHWAY GATA3 PATHWAY
STEM PATHWAY EPITHELIAL CELL SIGNALING IN HELICOBACTER
PARKISONS DISEASE CARDIAC EGF PATHWAY
STARCH AND SUCROSE METABOLISM NICOTINATE AND NICOTINAMIDE METABOLISM
EGF PATHWAY EDG1 PATHWAY
INSULIN PATHWAY PANTOTHENATE AND COA BIOSYNTHESIS
RENAL CELL CARCINOMA TOLL PATHWAY
WNT SIGNALING RENIN ANGIOTENSIN SYSTEM
AMINOACYL TRNA BIOSYNTHESIS ECM RECEPTOR INTERACTION
GALACTOSE METABOLISM PHOTOSYNTHESIS
O GLYCAN BIOSYNTHESIS NTH PATHWAY
ATM PATHWAY AMINOSUGARS METABOLISM
HUNTINGTONS DISEASE EICOSANOID SYNTHESIS
IL6 PATHWAY DORSO VENTRAL AXIS FORMATION
G1 TO S CELL CYCLE REACTOME NGF PATHWAY
FOCAL ADHESION SPRY PATHWAY
IGF1R PATHWAY ECM PATHWAY
UBIQUITIN MEDIATED PROTEOLYSIS MET PATHWAY
GLIOMA CDMAC PATHWAY
TPO PATHWAY ARF PATHWAY
G2 PATHWAY P38 MAPK PATHWAY
PORPHYRN AND CHLOROPHYLL METABOLISM CHOLERA INFECTION
GATA3 PATHWAY HYPERTROPHY MODEL
STEM PATHWAY WNT BETA CATENIN PATHWAY
PARKISONS DISEASE ERK PATHWAY
STARCH AND SUCROSE METABOLISM FMLP PATHWAY
EGF PATHWAY GLYCOLYSIS
INSULIN PATHWAY BLADDER CANCER
RENAL CELL CARCINOMA GATA3 PATHWAY
WNT SIGNALING EPITHELIAL CELL SIGNALING IN HELICOBACTER
AMINOACYL TRNA BIOSYNTHESIS CARDIAC EGF PATHWAY
GALACTOSE METABOLISM NICOTINATE AND NICOTINAMIDE METABOLISM
O GLYCAN BIOSYNTHESIS EDG1 PATHWAY
ATM PATHWAY PANTOTHENATE AND COA BIOSYNTHESIS
HUNTINGTONS DISEASE TOLL PATHWAY
IL6 PATHWAY RENIN ANGIOTENSIN SYSTEM
G1 TO S CELL CYCLE REACTOME ECM RECEPTOR INTERACTION
FOCAL ADHESION PHOTOSYNTHESIS
IGF1R PATHWAY NTH PATHWAY
UBIQUITIN MEDIATED PROTEOLYSIS AMINOSUGARS METABOLISM
GLIOMA EICOSANOID SYNTHESIS
TPO PATHWAY DORSO VENTRAL AXIS FORMATION
G2 PATHWAY NGF PATHWAY
SPRY PATHWAY
ECM PATHWAY
MET PATHWAY
CDMAC PATHWAY
ARF PATHWAY
P38 MAPK PATHWAY
CHOLERA INFECTION
HYPERTROPHY MODEL
WNT BETA CATENIN PATHWAY
ERK PATHWAY
FMLP PATHWAY
GLYCOLYSIS
IM CTLA4 PATHWAY NO IL12 PATHWAY
NO2 IL12 PATHWAY CTLA4 PATHWAY
TH1 TH2 PATHWAY IL12 PATHWAY
CSK PATHWAY TYPE I DIABETES MELLITUS
NKT PATHWAY NK CELL PATHWAY
COMPLIMENT PATHWAY TH1 TH2 PATHWAY
IL7 PATHWAY TNFR2 PATHWAY
PROTEASOME CSK PATHWAY
NK CELLS PATHWAY IL7 PATHWAY
TYPE I DIABETES MELLITUS AMI PATHWAY
AMP PATHWAY ANTIGEN PROCESSING AND PRESENTATION
ANTIGEN PROCESSING AND PRESENTATION NFKB PATHWAY
IL12 PATHWAY TUMOR NECROSIS FACTOR PATHWAY
LAIR PATHWAY PROTEASOME
TNFR2 PATHWAY NKT PATHWAY
CD PATHWAY T CELL SIGNAL TRANSDUCTION
TID PATHWAY DEATH PATHWAY
APOPTOSIS GENMAPP APOPTOSIS GENMAPP
NFKB PATHWAY CD40 PATHWAY
PROTEASOME PATHWAY RELA PATHWAY
T CELL SIGNAL TRANSDUCTION TOB1 PATHWAY
TUMOR NECROSIS FACTOR PATHWAY APOPTOSIS
CASPASE PATHWAY APOPTOSIS KEGG
APOPTOSIS T CELL RECEPTOR SIGNALING PATHWAY
NATURAL KILLER CELL MEDIATED CYTOTOXICITY CYTOKINE PATHWAY
TOB1 PATHWAY CASPASE PATHWAY
B CELL RECEPTOR SIGNALING PATHWAY TID PATHWAY
T CELL RECEPTOR SIGNALING PATHWAY IL2 PATHWAY
CYTOKINE PATHWAY NATURAL KILLER CELL MEDIATED CYTOTOXICITY
CELL ADHESION MOLECULES CASPASE CASCADE
DEATH PATHWAY LAIR PATHWAY
APOPTOSIS KEGG HEMATOPOIETIC CELL LINEAGE
TOLL LIKE RECEPTORS SIGNALING PATHWAY IL3 PATHWAY
PROTEASOME COMP PATHWAY
HEMATOPOIETIC CELL LINEAGE TCR PATHWAY
RELA PATHWAY B CELL RECEPTOR SIGNALING PATHWAY
IL2 PATHWAY STEM PATHWAY
MITOCHONDRIA PATHWAY MITOCHONDRIA PATHWAY
TOLL PATHWAY TOLL PATHWAY
B CELL ANTIGEN RECEPTOR STRESS PATHWAY
INFLAM PATHWAY IL2RB PATHWAY
IL2RB PATHWAY INFLAM PATHWAY
IL3 PATHWAY CYTOKINE CYTOKINE RECEPTOR INTERACTION
STEM PATHWAY PIP3 SIGNALING IN B LYMPHOCYTES
BCR SIGNALING PATHWAY TOLL LIKE RECEPTORS SIGNALING PATHWAY
HIVNEF PATHWAY 41B PATHWAY
TCR PATHWAY B CELL RECEPTOR SIGNALING PATHWAY
CASPASE CASCADE BCR SIGNALING PATHWAY
PIP3 SIGNALING IN B LYMPHOCYTES PROTEASOME
NTH PATHWAY CERAMIDE PATHWAY
41B PATHWAY APOPTOSIS
CCR5 PATHWAY JAX STAT SIGNALING PATHWAY
CYTOKINE CYTOKINE RECEPTOR INTERACTION EPO PATHWAY
INTERLEUKIN 4 PATHWAY NTH PATHWAY
STRESS PATHWAY CD40 PATHWAYMAP
IL1R PATHWAY CALCINEURIN NF AT SIGNALING
APOPTOSIS HIV NEP PATHWAY
EICOSANOID SYNTHESIS EICOSANOID SYNTHESIS
UBIQUITIN MEDIATED PROTEOLYSIS BCR PATHWAY
CELL CYCLE PATHWAY FC EPSILON RI SIGNALING PATHWAY
CALCINEURIN NF AT SIGNALING KERATINOCYTE PATHWAY
FC EPSILON RI SIGNALING PATHWAY AMINOACYL TRNA BIOSYNTHESIS
AMINOACYL TRNA BIOSYNTHESIS CELL CYCLE PATHWAY
JAK STAT SIGNALING PATHWAY B CELL RECEPTOR COMPLEXES
FAS PATHWAY IL8 PATHWAY
PTEN PATHWAY PROTEASOME PATHWAY CCRS PATHWAY
B CELL RECEPTOR COMPLEXES ATR BRCA PATHWAY
DNA REPLICATION REACTOME GHP PATHWAY
FFOLATEBIOSYNTHESIS FOLATE BIOSYNTHESIS
ATR BRCA PATHWAY PEPTIDE GPCRS
IL6 PATHWAY GLEEVEC PATHWAY
LEUKOCYTE TRANSENDOTHELIAL MIGRATION FAS PATHWAY
GLEEVEC PATHWAY ABC TRANSPORTERS GENERAL
TNFR PATHWAY INTERLEUKIN 4 PATHWAY
COMPLIMENT AND COAGULATION CASCADES IL4 RECEPTOR IN B LYPHOCYTES
BCR PATHWAY ADIPOCYTOKINE SIGNALING PATHWAY
EPO PATHWAY PROSTAGLANDIN AND LEUKOTRIENE METABOLISM
GH PATHWAY CDMAC PATHWAY
TPO PATHWAY TPO PATHWAY
AMINOACYL TRNA BIOSYNTHESIS HSP PATHWAY
PEPTIDES GPCRS TNFR1 PATHWAY
N GLYCAN BIOSYNTHESIS INOSITOL PHOSPHATE METABOLISM
ALZHEIMERS DISEASE ACUTE MYELOID LEUKEMIA
ACUTE MYELOID LEUKEMIA LEUKOCYTE TRANSENDOTHELIAL MIGRATION
GAQ PATHWAY IL1R PATHWAY
CD40 PATHWAYMAP IL1R ATROPHY
METHIONINEMETABOLISM OVARIAN INFERTILITY GENE
CPCR CLASS B SECRETIN LIKE PTEN PATHWAY
IL4 RECEPTOR IN B LYPHOCYTES
CERAMIDE PATHWAY
FCER1 PATHWAY
G1 TO S CELL CYCLE REACTOME
PARKINSONS DISEASE
CHEMICAL PATHWAY
CELL CYCLE KEGG
NARE INTERACTIONS IN VESICULAR TRANSPORT
ATM PATHWAY
NICOTINATE AND NICOTINAMIDE METABOLISM
KERATINOCYTE PATHWAY
HYPERTROPHY MODEL
ADIPOCYTOKINE SIGNALING PATHWAY
FML PATHWAY
OCITYOSELLUM DISODIUM CAMP CHEMOTAXIS
PEPTIDES PATHWAY
PANCREATIC CANCER
RAS SIGNALING PATHWAY
RAC PATHWAY
AMINOSUGARS METABOLISM
M CELL COMMUNICATION PTEN PATHWAY
HEPARAN SULFATE BIOSYNTHESIS DNA REPLICATION REACTOME
ECM RECEPTOR INTERACTION RIBOSOME
ALK PATHWAY TRANSLATION FACTORS
UCAL PAIN PATHWAY RNA POLYMERASE
STRIATED MUSCLE CONTRACTION DNA POLYMERASE
BASAL CELL CARCINOMA BASAL TRANSCRIPTION FACTORS
REGULATION OF THE ACTIN CYTOSKELETON BY IGF1 MTOPATHWAY
RHO GTPASES RNA REPLICATION REACTOME
TGFB SIGNALING PATHWAY CHOLESTEROL BIOSYNTHESIS
JDAC PATHWAY GLYCOSLPHYSPHATIDYLINOSITAL ANCHOR
HEDGEHOG SIGNALING PATHWAY ECM PATHWAY
INOSITOL PHOSPHATE METABOLISM UCAL PATHWAY
IGF1 PATHWAY EIF4 PATHWAY
ECM PATHWAY RIBOSOMAL PROTEINS
ERK5 PATHWAY CELL CYCLE KEGG
FOCAL ADHESION MTOR PATHWAY
EDG1 PATHWAY REGULATION OF THE ACTIN CYTOSKELETON BY RHO
CARM EER PATHWAY GTPASES
INSULIN PATHWAY GPASES
ETHER LIPD METABOLISM G1 TO S CELL CYCLE REACTOME
TEL PATHWAY MRNA PROCESSING REACTOME
NOTCH SIGNALING PATHWAY UBIQUITIN MEDIATED PROTEOLYSIS
WNT PATHWAY
ALK PATHWAY
G2 PATHWAY
TEL PATHWAY
CELL CYCLE
ATR BRCA PATHWAY
MSL PROSTAGLANDIN SYNTHESIS REGULATION NO2 IL12 PATHWAY
BAD PATHWAY CSK PATHWAY
LAIR PATHWAY LAIR PATHWAY
IL7 PATHWAY TOB1 PATHWAY
COMP PATHWAY CTLA4 PATHWAY
REMIN ANGIOTENSION SYSTEM AMP PATHWAY
AMP PATHWAY TH1 TH2 PATHWAY
CSK PATHWAY RENIN ANGIOTENSION SYSTEM
ERYTH PATHWAY PAR1 PATHWAY
ECM RECEPTOR INTERACTION NKT PATHWAY
TOB1 PATHWAY IL7 PATHWAY
COMPLEMENT AND COAGULATION CASCADES NK CELLS PATHWAY
CARDIAC EGF PATHWAY CD PATHWAY
HISTIDINE METABOLISM ERYTH PATHWAY
GATA3 PATHWAY HEMATOPOIETIC CELL LINEAGE
NDKDTNAMIN PATHWAY TYPE 1 DIABETES MELLITUS
GCRS PATHWAY IL3 PATHWAY
FOCAL ADHESION GLYCOSAMINOGLYCAN DEGRADATION
BETA ALANINE METABOLISM IL12 PATHWAY
INTRINSIC PATHWAY IL2 PATHWAY
PAR1 PATHWAY PROSTAGLANDIN SYNTHESIS REGULATION
ALK PATHWAY EICOSANOID SYNTHESIS
CALCINEURIN PATHWAY COMP PATHWAY
NTH PATHWAY CCR5 PATHWAY
CTLA4 PATHWAY GLYCAN STRUCTURES DEGRADATION
GCR PATHWAY CALCINEURIN PATHWAY
HEMATOPOIETIC CELL LINEAGE COMPLEMENT AND COAGULATION CASCADE
NO1 PATHWAY ECM RECEPTOR INTERACTION
VIP PATHWAY TCR PATHWAY
UCAL PAIN PATHWAY TNFR2 PATHWAY
EDG1 PATHWAY NO1 PATHWAY
IL3 PATHWAY INTRINSIC PATHWAY
IGF1 PATHWAY STATIN PATHWAY PHARMGK3
TCR PATHWAY ALKALOID BIOSYNTHESIS II
EICOSANOID SYNTHESIS LEUKOCYTE TRANSENDOTHELIAL MIGRATION
NK CELLS PATHWAY CELL ADHESION MOLECULES
VALINE AND ISOLEUCINE DEGRADATION INFLAM PATHWAY
PPARA PATHWAY MEF2D PATHWAY
SPRY PATHWAY IL2RB PATHWAY
HDAC PATHWAY WNT BETA CATENIN PATHWAY
STATIN PATHWAY FOCAL ADHESION
GLYCAN STRUCTURES DEGRADATION NTH PATHWAY
SMOOTH MUSCLE CONTRACTION BCR PATHWAY
TH1 TH2 PATHWAY BCR SIGNALING PATHWAY
ECM PATHWAY GLYCEROLIPID METABOLISM
PDGR PATHWAY HISTIDINE METABOLISM
INTEGRIN MEDIATED CELL ADHESION KEGG GATA3 PATHWAY
FATTY ACID METABOLISM PEPTIDE GPCRS
NKT PATHWAY PIP3 SIGNALING IN B LYMPHOCYTES
CXCR4 PATHWAY CYTOKINE CYTOKINE RECEOTIR INTERACTION
TOLL PATHWAY INOITOL PHOSPHATE METABOLISM
TNFR2 PATHWAY BAD PATHWAY
LEUKOCYTE TRANSENDOTHELIAL MIGRATION MONOAMINE GPCRS
PPAR SIGNALING PATHWAY NFKB PATHWAY
AMYOTROPHIC LATERAL SCLEROSIS ECM PATHWAY
GLYCEROLIPID METABOLISM 1 AND 2 METHYLNAPHTHALENE DEGRADATION
GLYCOSPHINGOLIPID METABOLISM B CLL RECEPTOR SIGNALING PATHWAY
MTOR PATHWAY VIP PATHWAY
NICOTINATE AND NICOTINAMIDE METABOLISM CARDIAC EGF PATHWAY
PGC1 PATHWAY IL4 RECEPTOR IN B LYPHOCYTES
MET PATHWAY GH PATHWAY
WNT BETA CATENIN PATHWAY NATURAL KILLER CELL MEDIATED CYTOTOXICITY
TPO PATHWAY CYTOTOXICITY
BUTANOATE METABOLISM RAC1 PATHWAY
TGF BETA SIGNALING PATHWAY EDG1 PATHWAY
GSK3 PATHWAY BETA ALANINE METABOLISM
GLYCOSAMINOGLYCAN DEGRADATION UCAL PAIN PATHWAY
PROPNOATE METABOLISM GPCR PATHWAY
CELL COMMUNICATION ERK1 ERK2 MAPK PATHWAY
OVARIAN INFERTILITY GENES INTERGRIN MEDIATED CELL ADHESION KEGG
VALINE LEUCINE AND ISOLEUCINE DEGRADATION ARACHIDONIC ACID METABOLISM
BILE ACID BIOSYNTHESIS ABC TRANSPORTERS GENERAL
IL5 PATHWAY RHO PATHWAY
BCR SIGNALING PATHWAY JAK STAT SIGNALING PATHWAY
CELL ADHESION MOLECULES HDAC PATHWAY
B CLL RECEPTOR SIGNALING PATHWAY BILE ACID BIOSYNTHESIS
BCR PATHWAY TOLL PATHWAY
PTEN PATHWAY BPA PATHWAY
MCAL PAIN PATHWAY NDKDTHAMIN PATHWAY
1 AND 2 METHYLNAPHTHALENE DEGRADATION SMOOTH MUSCLE CONTRACTION
GFCDB CLASS B SECRETIN LIKE CALCIUM SIGNALING PATHWAY
IL2 PATHWAY GLYCEROLIPID METABOLISM
MELANOMA ANTIGEN PROCESSING AND PRESENTATION
INTERGRIN PATHWAY ADIPOCYTOKINE SIGNALING PATHWAY
ALKALOID BIOSYNTHESIS II
ETHER LIPID METABOLISM
NUCLEAR RECEPTORS CK1 PATHWAY
TYPE I DIABETES MELLITUS
ND2IL12 PATHWAY
RHO PATHWAY
CYTOKINE CTYOKINE RECEPTOR INTERACTION
BLOOD CLOTTING CASCADE
INFLAM PATHWAY
DICYTOSTELIUM DISODIUM CAMP
CHEMOTAXIS PATHWAY
DIFFERENTIATION PATHWAY IN PC12 CELLS
LAR CHOLESTEROL BIOSYNTHESIS GLUTATHIONE METABOLISM
PENTOSE AND GLUCURONATE PENTOSE AND GLUCURONATE INTERCONVERSIONS
INTERCONVERSIONS CHOLESTEROL BIOSYNTHESIS
BIOSYNTHESIS OF STEROIDS TYROSINE METABOLISM
TYROSINE METABOLISM BIOSYNTHESIS OF STEROIDS
GAMMA HEXACHLOROCYCLOHEXANE PORPHYRIN AND CHLOROPHYLL METABOLISM
DEGRADATION ANDROGEN AND ESTROGEN METABOLISM
PORPHYRIN AND CHLOROPHYLL METABOLISM GLYCOSPHINGOLIPID METABOLISM
PHENYLALANINE METABOLISM TYPE I SECRETION SYSTEM
CHREB PATHWAY GAMMA HEXACHLOROCYCLOHEXANE DEGRADATION
GLUTATHIONE METABOLISM FLAGELLAR ASSEMBLY
1 AND 2 METHYLNAPHTHALENE DEGRADATION CITRATE CYCLE TCA CYCLE
GLYCOSPHINGOLIPID METABOLISM PHENYLALANINE METABOLISM
PORPHYRIN AND CHLOROPHYLL METABOLISM ATP SYNTHESIS
ANDROGEN AND ESTROGEN METABOLISM PHOTOSYNTHESIS
PHENILALANINE METABOLISM STARCH AND SUCROSE METABOLISM
CITRATE CYCLE TCA CYCLE PORPHYRIN AND CHLOROPHYLL METABOLISM
METABOLISM OF XENOBIOTICS BY CYTOCHROME ARGININE AND PROLINE METABOLISM
P450 METABOLISM OF XENOBIOTICS BY CYTOCHROME P450
EICOSANOID SYNTHESIS FRUCTOSE AND MANNOSE METABOLISM
VALINE LEUCINE AND ISOLEUCINE DEGRADATION CARBON FIXATION
GLYCAN STRUCTURES DEGRADATION GLYCAN STRUCTURES DEGRADATION
ANCHOR BIOSYNTHESIS COMP PATHWAY
BUTANOATE METABOLISM PARKINSONS DISEASE
ARGININE AND PROLINE METABOLISM PANTOTHENATE AND COA BIOSYNTHESIS
GLUTATHIONE METABOLISM FATTY ACID METABOLISM
CK1 PATHWAY RENIN ANGIOTENSIN SYSTEM
CITRATE CYCLE TYROSINE METABOLISM
PANTOTHENATE AND COA BIOSYNTHESIS GLUTAMATE METABOLISM
PROPANOATE METABOLISM ALANINE AND ASPARTATE METABOLISM
FATTY ACID METABOLISM EICOSANOID SYNTHESIS
GLUTAMATE METABOLISM GLYCOSAMINOGLYCAN DEGRADATION
N GLYCAN BIOSYNTHESIS PHENILALANINE METABOLISM
HISTIDINE METABOLISM CREB PATHWAY
BILE ACID BIOSYNTHESIS CK1 PATHWAY
GLUTAMATE METABOLISM MONOAMINE GPCRS
TRYPTOPHAN METABOLISM PHINGOLIPID METABOLISM
ALANINE AND ASPARTATE METABOLISM P53 HYPOXIA PATHWAY
PPAR SIGNALING PATHWAY TRYPTOPHAN METABOLISM
CREB PATHWAY NICOTINATE AND NICOTINAMIDE METABOLISM
ATP SYNTHESIS GLUTAMATE METABOLISM
FLAGELLAR ASSEMBLY UCAL PAIN PATHWAY
TYPE III SECRETION SYSTEM KREBS TCA CYCLE
SPRY PATHWAY OXIDATIVE PHOSPHORYLATION
ERYTH PATHWAY HISTIDINE METABOLISM
PHOTOSYNTHESIS GALACTOSE METABOLISM
MCAL PAIN PATHWAY PENTOSE PHOTPHATE PATHWAY
FRUCTOSE AND MANNOSE METABOLISM N GLYCAN BIOSYNTHESIS
GLYCOSAMINOGLYCAN DEGRADATION LINOLEIC ACID METABOLISM
SPHINGOLIPID METABOLISM BLOOD CLOTTING CASCADE
GLUTAMATE METABOLISM ARACHONDIC ACID METABOLISM
GLYCEROLIPID METABOLISM MCAL PAIN PATHWAY
SNARE INTERACTIONS IN VESICULARE TRANSPORT ERYTH PATHWAY
SELENAMINO ACID METABOLISM ABO TRANSPORTERS GENERAL
PENTOSE PHOSPHATE PATHWAY BUTANOATE METABOLISM
NO1 PATHWAY O GLYCAN BIOSYNTHESIS
STARCH AND SUCROSE METABOLISM PPAR SIGNALING PATHWAY
CYSTEINE METABOLISM VALINE LEUCINE ISOLEUCINE DEGRADATION
GATA3 PATHWAY NO1 PATHWAY
ABO TRANSPORTERS GENERAL COMPLEMENT AND COAGULATION CASCADES
UREA CYCLE AND METABOLISM OF AMINO
GROUPS
OXIDATIVE PHOSPHORLATION
KREBS TCA CYCLE
AMINOSUGARS METABOLISM
ARACHONDIC ACID METABOLISM
NAPHTHALENE AND ANTHRACENE DEGRADATION
OXIDATIVE PHOSPHORYATION
UNC DNA REPLICATION REACTOME
ATR BRC PATHWAY
DNA POLYMERASE
CELL CYCLE KEGG
CELLCYCLE PATHWAY
G1 TO S CELL CYCLE REACTOME
CELL CYCLE
B1 PATHWAY
G1 AND S PHASES
BASAL TRANSCRIPTION FACTORS
RNA TRANSCRIPTING REACTOME
AMINOACYL TRNA BIOSYNTHESIS
ARNA POLYMERASE
PYRIMIDINE METABOLISM
CARM ER PATHWAY
PYRIMIDINE METABOLISM
ALANINE AND ASPARTATE METABOLISM
G2 PATHWAY
P53 PATHWAY
SELENOAMINO ACID PATHWAY
GLUTAMATE METABOLISM
PENTOSE PHOSPHATE PATHWAY
Bold-shared between training and validation set
TABLE 5
Cell Line Culture Conditions
Cell Line Site OF Origin Tumor Type BRCA1 Mutation Source Media
BT20 PB IDC ATCC EMEM 10% FBS
BT549 [B ODC ATCC RPMI 10% FBS
CAL120 PE AC DSMZ DMEM 10% FBS
CAL148 PE AC DSMZ DMEM 20% + 2 Mm
L-glutamine + 1 μg/ml
EGF
CAL851 PE IGA DSMZ DMEM 10% FBS +
2 Mm L-glutamine
DU4475 CN DC ATCC RPMI 10% FBS
HCC1143 PB IDC ATCC RPMI 10% FBS
HCC1187 PB DC ATCC RPMI 10% FBS
HCC1395 PB DC ATCC RPM″ 10% FBS
HCC1599 PB DC ATCC RPMI 10% FBS
HCC1806 PB ASCC ATCC RPMI glutamax 10% FBS
HCC1937 PB DC X ATCC RPM (glutamax 10% FBS
HCC38 PB DC ATCC RPMI 10% FBS
HCC70 PB DC ATCC RPMI 10% FBS
HDQP1 PB IDC DSMZ DMEM 10% FBS
HS578T PB CS ATCC DMEM 10% FBS
MDAMB157 PB MC ATCC DMEM 10% FBS
MDAMB231 PE IDC ATCC DMEM 10% FBS
MDAM436 PE IDC X ATCC DMEM 10% FBS
MDAM453 PE AC ATCC DMEM 10% FBS
MDAM468 PE DC ATCC DMEM 10% FBS
MFM223 PE AC DSMZ MEM (with Earle's
sals) + 15% FBS + 2 Mm
L-glutamine + 1x insulin-
transferrin-sodium s
MT3 PE C DSMZ RPMI 10% FBS
SUM149PT PB INF X Asterand Hams F12 5% FBS +
1 μg/ml hydrocortisone +
5 μg/ml insulin
SUM159PT PB ANC Asterand Hams F12 5% FBS +
1 μg/ml hydrocortisone +
5 μg/ml insulin
SUM185 PE DC Asterand Hams F12 5% FBS +
1 μg/ml hydrocortisone +
5 μg/ml insulin
SW527 PE ATCC DMEM 10% FBS
Tumour Type (AC, adenocarcinoma; ANC, anaplastic carcinoma; ASCC, acantholytic squamous cell carcinoma; CS, Carcinosarcoma; DC ductal carcinoma; C, carcinoma; CS, arcinosarcoma; IDC, invasive d; Isolation (PB, primary breast; PE, pleural effusion; CN cutaneous nodule)
TABLE 6A
Gene Categories
BLI BL2 IM M MSL LAR
ACTG2 ALXO5 ADAMDEC1 ACTA2 ABCA8L ABCA12
ADAMDEC1 CLCA2 AIM2 ACTG2 ABCB1 AKAP12
ANP32E COL5A1 ALOX5 BCL11A ACTA2 AKR1B10
APOCBEC3A CTSC ANP32E BGN E ACTG2 AKR1D1
ATR3 CXCL12 APOBEC3A CAV1 E AKAP12 ALCAM G
AURKA A DHCR24 APOBEC3G CAV2 E ALDHA1 L ALDH3B2 G
AURKB A EGFR D BCL2AA CCND2 E ALOX5 ALOX15B
AZGP1 EPHA2 D BIRC3 COL3A1 E AOC3 APOD G
BCL11A GALNT10 BTK3A3 COL5A1 E APOBEC3G AR
BIRC5 GBP2 BYK I COL5A2 E APOD AZGP1
BUB1 A GPR87 C1QA COL6A3 E AZGP1 BLVRB
CCL5 HTRA1 C1QB CTNNB1 E BCL2 C4B
CCNA2 A MET D C4B DKK2 E BGN E CADPS2
CENPA A MME CASP1 DKK3 E BMP2 E CLCA2
CENPFA PYCARD CCL19 H EN1 C4B CLDN8
CHEK1 C RSAD2 CCL3 H EPHB3 CAV1 E COL5A1
CKAP2 S100A7 CCL4 H FBN1 E CAV2 E COL6A3
COBL S100A8 CCL5 H FOXC1 CCND2 E CRAT
CTSS S100A9 CCL8 H FZD4 E CCR2 H CUX2
CXCL10 TNFSF10 CCR1 H GNG11 CD2 DHCR24 G
CXCL11 TP63 CCR2 H GPR161 CD37 DHRS2
CXCL13 CCR5 H HTRA1 CD3D EAR2
CXCL9 CCR7 H IGF1 E CD48 FAH
EN1 CD2 K IGF2 E CD52 FASN G
EPHB3 CD37 K IGFBP4 CD69 FBN1
EXO1 C CD38 K IGFBP5 CD74 FKBP5 G
FANCA C CD3D K KCNK5 CKAP2 FMO5
FANCG C CD48 K LAMB1 COL3A1 E FOXA1 G
GBP1 CD52 K MFGE8 COL3A1 E G6PD
IFI44L CD69 K MIA COL5A2 E GALNT10
IFIH1 CD74 K MICALL1 COL6A3 E GALNT7
IGKC CD8A K MMP2 E CORO1A GGT1
IGKV1D-13 CKAP2 NOTCH1 E CSF2RB GGTLC3
IGKV4-1 CORO1A PDGFRA CTNNB1 E HGD
ITPKB CSF2RB PDGFRB CXCL12 H HMGCS2
IVD CTSC PMP22 CXCL13 H HSD17B11
LOC100130100 CTSS ROPN1 CYTIP HTRA1
LOC339562 CXCL10 H S100A1 DKK2 E INPP4B
LOC652493 CXCL11 H SCRG1 DKK3 E IQGAP2
MCM10 A CXCL13 H SFPR4 DPYD KCNMA1
MDC1 C CXCL14 H SMAD6 E ENG KMO
MFGE8 CXCL9 H SMAD7 E EPAS1 KRT18 G
MIA CXCR4 H SNAI2 EVI2A KYNU
MICALL1 CXCR6 H SOX10 EVI2B LRRC17
MSH2 C CYBB SPARC E FBN1 E MLPH
MUC5B CYTIP SPOCK1 FCFL2 MSX2
MYBL1 DDX60 SRPX FGL2 PIP G
MYC B DPYD SYNM FYB POU2AF1
NBN C EN1 TAGLN E FZD4 E RARRES3
NRAS C EVI2A TCF4 E GALNT10 S100A7
NTN3 EVI2B TCF7L2 E GALNT7 S100A8
PADI2 FGL2 TGFB1 E GIMAP6 S100A9
PLK1 A FYB TGFBIL1 E GNG11 SERHL2
PMP22 GBP1 TGFB2 E GZMA SIDT1
POU2AF1 GBP2 TGFB3 E HCLS1 SPARCL1
PRC1 A GIMAP6 TGFBR1 E HLA-DMZ SPDEF G
PSBM9 GNLY TGFBR2 E HLA-DPB1 SPOCK1
RAD21A GZMA TGFBR3 E HLA-DRA TARP
RAD51C GZMB TRPS1 HLA-DRB1 TFAP2B
RAD54BPC HCLS1 TWIST1 E HOXA10MEIS1 TNFSF10
ROPN1 HCP5 WWP2 HOXA5 TP53G1
RRSAD2 HERC5 ZEB1 E HSD17B11 TRGC2
RTP4 HLA-DMA ZEB2 E HTRA1 TRGV9
S100A1 HLA-DPB1 IGF1 E TRIM36
SCRG1 HLA-DRA IGF2 E UGT2B38
SOX10 HLA-DRB1 IGFBP4 XBP1 G
SYNM IDO1 IGFBP5 ZBTB16
TAP1 IF135 IGKC
TCF7L1 IF144L IGKV1D-13
TP53BP2 C IFIH1 IGKV4-1
TRPS1 IGKC IL23A
TTK B IFNG J IL2RG
UBD B IGKV1D-13 IQGAP2
IGKV4-1 IRF8
IL10RA J ITPKB
IL15RA J IVD
IL16 J KDR
IL18 J LAMB1
IL18R1 J LCK
IL2RN J LHFP
IL23A J LOC100130100
IL2RA J LOC652493
IL2RB J LRRC17
IL2RG J LTB
IL4 J MEIS2 L
IL6 J MEOX1 L
IL7 J MEOX2 L
IRF1 J MMP2
IRF7 J MSX1ITGAV
IRF8 J NGFR
ITK NOTCH1
IVD NT5E
JAK1 J NTN3
KYNU
LAMP3
LCK PDGFRA
LOC100130100 PDGFRB
LOC339562 PER1
LOC652493 PLAC8
LTB PMP22
LYN I POU2AF1
NPKB1 I PROCR L
NKG7 PTPRC
NTN3 RAC2 E
OAS1 ROPN1
OAS2 SAMSN1
OASL SELL
PLAC8 SFPR4
POU2AF1 SMAD6 E
PSMB9 SMAD7 E
PTPRC SNAI2 E
PYCARD SPARC E
RAC2 I SPARCL1 E
RARRES3 SPOCK1
RELB SRGN
RSAD2 SRPX
RTP4 SYNM
S100A8 TAGLN E
SAMSN1 TCF4 E
SELL TCF7L2 E
SLAMF7 TEK
SNX10 TERF2IP
SRGN TGFB1 E
STAT1 I TGFB1L1 E
STAT4 I TGFB2 E
STAT5A I TGFB3 E
TAP1 TGFBR1 E
TARP TGFBR2 E
TFEC TGFBR3 E
TNFAIP2 THBS4
TNFSF17 THY1 L
TNFSF10 TIE1
TRAC TNFRSF17
TRGC2 TRAC
TRGV9 TRIM22
TRIM22 TWIST1 E
UBD VCAM1
UB2EL6 VIM E
WARS XBP1
ZBTB16
ZEB1 E
ZEB2 E
A Cell division
B Cell proliferation
C DNA damage
D Growth factor
E ECM/TGFB
F WNT
G AR responsive
H Chemokine
I Immune signal transduction
J Cytokine signaling
K Surface antigen
L mesenchymal stem genes
TABLE 6B
TNBC Subtype Mutations
BLI BL2 IM M MSL LAR
TOP2B BRCA1 TP53 APC APC MAP3K1
ATM BRCA2 STAT4 BRAF BRAF PIK3CA
ATR CDKN2A STAT1 CTNNB1 BCRA RB1
BRCA1 PTCH1 RET FGFR1 CDKN2A TP53
BRCA2 PTCH2 NFKBIA GLI1 CTNNB1
CAMK1G PTEN MAP2K4 HRAS FGFR1
CDKN2A RB1 HUWE1 KRAS HRAS
CLSPN RET BRAF NOTCH1 KRAS
CTNND1 TP53 NOTCH4 NF1
HDAC4 UTX PIK3CA NF2
MAPK13 PTEN PDGFRA
MDC1 RB1 PDGFRB
NOTCH1 TP53 PIK3CA
PTEN TP53
RB1
SMAD4
SMARCAL1
STAT4
TIMELESS
TP53
UTX
TABLE 6C
Cell Line Source
Cell Line Subtype Source Website Product No.
HCC1143 BL1 ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ CRL-2321
tabid/452/Default.aspx?ATTCNum=CRL-2321&Template=cellBiology
HCC1599 BL1 ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ CRL-2331
tabid/452/Default.aspx?ATTCNum=CRL-2331&Template=cellBiology
HCC38 BL1 ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ CRL-2314
tabid/452/Default.aspx?ATTCNum=CRL-2314&Template=cellBiology
HCC2157 BL1 ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ CRL-2340
tabid/452/Default.aspx?ATTCNum=CRL-2340&Template=cellBiology
HCC1937 BL1 ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ CRL-2336
tabid/452/Default.aspx?ATTCNum=CRL-2321&Template=cellBiology
HCC3153 BL1 NA NA NA
MDA-MB-468 BL1 ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ HTB-132
tabid/452/Default.aspx?ATTCNum=HTB-132&Template=cellBiology
CAL-851 BL2 DSMZ http://www.dsmz.de/catalogus/details/culture/ACC- ACC-440
440.html?tx_dsmzresources_pi5[returnPid]=192
HCC70 BL2 ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ CRL-2315
tabid/452/Default.aspx?ATTCNum=CRL-2315&Template=cellBiology
HDQP1 BL2 DSMZ http://www.dsmz.de/catalogus/details/culture/ACC- ACC-494
494.html?tx_dsmzresources_pi5[returnPid]=192
SUM149PT BL2 Asterand http://www.asterand.com/Asterand/human-tissues/149PT.htm SUM-149PT
HCC1806 BL2 ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ CRL-2335
tabid/452/Default.aspx?ATTCNum=CRL-2335&Template=cellBiology
DU4475 IM ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ HTB-123
tabid/452/Default.aspx?ATTCNum=HTB-123&Template=cellBiology
HCC1187 IM ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ CRL-2322
tabid/452/Default.aspx?ATTCNum=CRL-2322&Template=cellBiology
CAL-148 LAR DSMZ http://www.dsmz.de/catalogus/details/culture/ACC- ACC-460
460.html?tx_dsmzresources_pi5[returnPid]=192
MFM223 LAR DSMZ http://www.dsmz.de/catalogus/details/culture/ACC- ACC-422
422.html?tx_dsmzresources_pi5[returnPid]=192
SUM185PE LAR Asterand http://www.asterand.com/Asterand/human-tissues/185PE.htm SUM-185PE
MDA-MB-453 LAR ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ HTB-131
tabid/452/Default.aspx?ATTCNum=HTB-131&Template=cellBiology
HCC2185 LAR NA NA NA
BT549 M ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ HTB-122
tabid/452/Default.aspx?ATTCNum=HTB-122&Template=cellBiology
CAL-120 M DSMZ http://www.dsmz.de/catalogus/details/culture/ACC- ACC-459
459.html?tx_dsmzresources_pi5[returnPid]=192
CAL-51 M DSMZ http://www.dsmz.de/catalogus/details/culture/ACC- ACC-302
302.html?tx_dsmzresources_pi5[returnPid]=192
HS578T MSL ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ HTB-126
tabid/452/Default.aspx?ATTCNum=HTB-126&Template=cellBiology
MDA-MB-157 MSL ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/
tabid/452/Default.aspx?ATTCNum=HTB-24&Template=cellBiology HTB-24
MDA-MN-436 MSL ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/
tabid/452/Default.aspx?ATTCNum=HTB-130&Template=cellBiology HTB-130
SUM159PT MSL Asterand http://www.asterand.com/Asterand/human-tissues/159PT.htm SUM-159PT
MDA-MB-231 MSL ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ HTB-26
tabid/452/Default.aspx?ATTCNum=HTB-26&Template=cellBiology
BT20 ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ HTB-19
tabid/452/Default.aspx?ATTCNum=HTB-19&Template=cellBiology
HCC1395 ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ CRL-2324
tabid/452/Default.aspx?ATTCNum=CRL-2324&Template=cellBiology
SW527 ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ CRL-7940
tabid/452/Default.aspx?ATTCNum=CRL-7940&Template=cellBiology
MCF10A ATCC http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/ CRL-10317
tabid/452/Default.aspx?ATTCNum=CRL-10317&Template=cellBiology
The teachings of all patents, published applications and references cited herein are incorporated by reference in their entirety.
While this invention has been particularly shown and described with references to example embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims (17)

What is claimed is:
1. A method of determining and treating a triple negative breast cancer (TNBC) subtype in a human individual in need thereof comprising:
a) determining expression of at least ABCA12, ADAMDEC1, AKR1B10, ALCAM, ALDH3B2, ALOX15B, ALOX5, ANP32E, APOBEC3G, APOD, AR, AZGP1, BCL11A, BCL2A1, BIRC3, C1QA, CADPS2, CCL5, CCR2, CD2, CD38, CLCA2, COBL, COL5A1, COL5A2, COL6A3, CRAT, CTSC, CTSS, CXCL10, CXCL11, CXCL13, DDX60, DHCR24, DHRS2, EAF2, EN1, EPHB3, FASN, FGL2, FOXA1, FOXC1, FZD4, G6PD, GALNT10, GBP1, GBP2, GGT1, GGTLC3, GNLY, GPR161, GPR87, GZMA, GZMB, HGD, HMGCS2, HTRA1, IDO1, IGKC, IL15, INPP4B, IRF1, ITPKB, KCNK5, KCNMA1, KMO, KRT18, KYNU, LAMP3, LOC652493, LTB, MCM10, MFGE8, MIA, MICALL1, MLPH, MME, MMP2, MSX2, MUC5B, MYBL1, NKG7, NTN3, OAS1, PADI2, PIP, PLACE, PSMB9, PTPRC, RARRES3, ROPN1, S100A1, S100A7, S100A8, S100A9, SCRG1, SELL, SEMA4D, SERHL2, SIDT1, SOX10, SPDEF, STAT1, SYNM, TAP1, TCF7L1, TFAP2B, TGFBI, THY1, TNFSF10, TP53BP2, TP53TG1, TP63, TRIM36, TRPS1, UBD, UBE2L6, UGT2B28, WARS, and XBP1 genes in one or more TNBC cells of the individual;
b) comparing the expression of the genes in the TNBC cells with the expression of the genes in a control;
c) determining an increased expression of at least one of the genes in the TNBC cells as compared to the expression of the genes in the control;
d) determining if the TNBC subtype is TNBC IM or TNBC LAR from the increased expression of at least one of the genes; and
e) administering a treatment protocol for the individual based on the TNBC subtype;
wherein determining that the TNBC is the TNBC IM subtype includes determining increased expression of one or more genes selected from the group consisting of BCL2A1, BIRC3, C1QA, CD38, DDX60, GNLY, GZMB, IDO1, IL15, IRF1, LAMP3, NKG7, SEMA4D, STAT1, TAP1, UBE2L6, and WARS in the TNBC cells compared to the control;
wherein determining that the TNBC is the TNBC LAR subtype includes determining increased expression of one or more genes selected from the group consisting of ABCA12, AKR1B10, ALCAM, ALDH3B2, ALOX15B, AR, CADPS2, CRAT, DHRS2, EAF2, FASN, FOXA1, G6PD, GGTLC3, HGD, HMGCS2, INPP4B, KCNMA1, KMO, KRT18, MLPH, MSX2, PIP, SERHL2, SIDT1, SPDEF, TFAP2B, TP53TG1, TRIM36, and UGT2B28 in the TNBC cells compared to the control;
wherein the treatment protocol for the TNBC IM subtype includes treating the individual with an immune checkpoint inhibitor and the treatment protocol for the TNBC LAR subtype includes treating the individual with an AR antagonist alone or in combination with PI3K inhibitors; and
wherein the control is selected from the group consisting of corresponding cells from one or more individuals that do not have TNBC, non-TNBC cells from the individual, a standard control established by assaying individuals that do not have TNBC, and combinations thereof.
2. The method of claim 1 wherein the TNBC cells are selected from the group consisting of epithelial cells, mesenchymal cells, immune cells, and combinations thereof.
3. The method of claim 1 wherein the control is the non-TNBC cells from the individual.
4. The method of claim 1 wherein the non-cancer cell is the corresponding cells from one or more individuals that do not have TNBC.
5. The method of claim 1 wherein determining that the TNBC is the TNBC IM subtype includes selectively determining an increased expression of a gene combination comprising CCL19, CCL3, CCL4, CCL5, CCL8, CCR1, CCR2, CCR5, CCR7, CD2, CD37, CD38, CD3D, CD48, CD52, CD69, CD74, and CD8A in the TNBC cells compared to the control.
6. The method of claim 1 wherein determining that the TNBC is the TNBC IM subtype further comprises detecting one or more mutated genes.
7. The method of claim 6 wherein the one or more mutated genes are selected from the group consisting of TP53, CTNNA1, DDX18, HUWE1, NFKBIA, APC, BRAF, MAP2K4, RB1, STAT4, STAT1, RET, and combinations thereof.
8. The method of claim 1 wherein the TNBC IM subtype is detected in the individual and the individual is treated with one or more immune checkpoint inhibitors.
9. The method of claim 1 wherein determining that the TNBC is the TNBC LAR subtype includes selectively detecting increased expression of a gene combination comprising AR, DHCR24, ALCAM, GATA2, GATA3, IDIH1, IDIH2, CDH11, ERBB3, CUX2, FGFR4, HOPX, FASN, FKBP5, APOD, PIP, SPDEF, CLDN8, FOXA1, KRT18, and XBP1 in the TNBC cells compared to the control.
10. The method of claim 1 wherein determining that the TNBC is the TNBC LAR subtype further comprises detecting one or more mutated genes.
11. The method of claim 10 wherein the one or more mutated genes are selected from the group consisting of PIK3CA, CDH1, PTEN, RB1, TP53, MAP3K1, and combinations thereof.
12. The method of claim 1 wherein the TNBC LAR subtype is detected in the individual and the individual is treated with one or more drugs that inhibit androgen receptor (AR).
13. The method of claim 12 wherein the one or more drugs that inhibit AR are selected from the group consisting of bicalutamide, MVD3100, abiraterone, and combinations thereof.
14. The method of claim 12 further comprising treating the individual with a PI3K inhibitor a PI3K/mTOR inhibitor or a combination thereof.
15. The method of claim 14 wherein the PI3K inhibitor is BKM-120, GDC0941 or a combination thereof, and the PI3K/mTOR inhibitor is NVP-BEZ235, GDC0980 or a combination thereof.
16. The method of claim 12 further comprising treating the individual with an inhibitor of HSP90.
17. The method of claim 16 wherein the inhibitor of HSP90 is DMAG.
US16/746,721 2011-11-18 2020-01-17 Markers of triple-negative breast cancer and uses thereof Active US11788147B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/746,721 US11788147B2 (en) 2011-11-18 2020-01-17 Markers of triple-negative breast cancer and uses thereof
US18/381,126 US20240158865A1 (en) 2011-11-18 2023-10-17 Markers of Triple-Negative Breast Cancer and Uses Thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161561743P 2011-11-18 2011-11-18
PCT/US2012/065724 WO2013075059A1 (en) 2011-11-18 2012-11-17 Markers of triple-negative breast cancer and uses thereof
US201414358330A 2014-05-15 2014-05-15
US16/746,721 US11788147B2 (en) 2011-11-18 2020-01-17 Markers of triple-negative breast cancer and uses thereof

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2012/065724 Continuation WO2013075059A1 (en) 2011-11-18 2012-11-17 Markers of triple-negative breast cancer and uses thereof
US14/358,330 Continuation US20140303133A1 (en) 2011-11-18 2012-11-17 Markers of Triple-Negative Breast Cancer And Uses Thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/381,126 Division US20240158865A1 (en) 2011-11-18 2023-10-17 Markers of Triple-Negative Breast Cancer and Uses Thereof

Publications (2)

Publication Number Publication Date
US20200224282A1 US20200224282A1 (en) 2020-07-16
US11788147B2 true US11788147B2 (en) 2023-10-17

Family

ID=47324427

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/358,330 Abandoned US20140303133A1 (en) 2011-11-18 2012-11-17 Markers of Triple-Negative Breast Cancer And Uses Thereof
US16/746,721 Active US11788147B2 (en) 2011-11-18 2020-01-17 Markers of triple-negative breast cancer and uses thereof
US18/381,126 Pending US20240158865A1 (en) 2011-11-18 2023-10-17 Markers of Triple-Negative Breast Cancer and Uses Thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/358,330 Abandoned US20140303133A1 (en) 2011-11-18 2012-11-17 Markers of Triple-Negative Breast Cancer And Uses Thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/381,126 Pending US20240158865A1 (en) 2011-11-18 2023-10-17 Markers of Triple-Negative Breast Cancer and Uses Thereof

Country Status (5)

Country Link
US (3) US20140303133A1 (en)
EP (1) EP2780469A1 (en)
AU (1) AU2012340186A1 (en)
CA (1) CA2856295A1 (en)
WO (1) WO2013075059A1 (en)

Families Citing this family (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108192972B (en) 2010-10-06 2022-09-09 生物医学研究机构基金会 Methods for diagnosis, prognosis and treatment of breast cancer metastasis
EP2650682A1 (en) 2012-04-09 2013-10-16 Fundació Privada Institut de Recerca Biomèdica Method for the prognosis and treatment of cancer metastasis
CA2894153A1 (en) * 2012-12-07 2014-06-12 The General Hospital Corporation Combinations of a pi3k/akt inhibitor compound with an her3/egfr inhibitor compound and use thereof in the treatment of a hyperproliferative disorder
US9551036B2 (en) * 2013-02-25 2017-01-24 Whitehead Institute For Biomedical Research Metabolic gene mesenchymal signatures and uses thereof
US10323285B2 (en) 2013-09-09 2019-06-18 Nantomics, Llc Proteomics analysis and discovery through DNA and RNA sequencing, systems and methods
JP2016540726A (en) * 2013-10-04 2016-12-28 シグナル ファーマシューティカルズ,エルエルシー TOR kinase inhibitor in prevention or treatment of cancer characterized by gene mutation
EP3074532A1 (en) * 2013-11-28 2016-10-05 Stichting Het Nederlands Kanker Instituut- Antoni van Leeuwenhoek Ziekenhuis Methods for molecular classification of brca-like breast and/or ovarian cancer
WO2015089402A1 (en) * 2013-12-12 2015-06-18 The University Of Chicago Methods and compositions related to hsp90 inhibitors and breast cancer
WO2015160882A1 (en) 2014-04-16 2015-10-22 Signal Pharmaceuticals, Llc SOLID FORMS COMPRISING 7-(6-(2-HYDROXYPROPAN-2YL) PYRIDIN-3-YL)-1-(TRANS)-4-METHOXYCYCLOHEXYL)-3, 4-DIHYDROPYRAZINO[2,3-b] PYRAZIN-2(1H)-ONE, AND A COFORMER, COMPOSITIONS AND METHODS OF USE THEREOF
US9545417B2 (en) 2014-05-01 2017-01-17 The Johns Hopkins University Methods of inhibiting cancer stem cells with HMGA1 inhibitors
KR20170005419A (en) * 2014-05-21 2017-01-13 에프. 호프만-라 로슈 아게 Methods of treating pr-positive, luminal a breast cancer with pi3k inhibitor, pictilisib
CN108064311B (en) 2014-10-24 2022-10-28 皇家飞利浦有限公司 Medical prognosis and prediction of treatment response using multiple cell signaling pathway activities
JP6415712B2 (en) 2014-10-24 2018-10-31 コーニンクレッカ フィリップス エヌ ヴェKoninklijke Philips N.V. Assessment of TGF-β cell signaling pathway activity using mathematical modeling of target gene expression
EP3210144B1 (en) * 2014-10-24 2020-10-21 Koninklijke Philips N.V. Medical prognosis and prediction of treatment response using multiple cellular signaling pathway activities
SG11201707067UA (en) * 2015-03-12 2017-09-28 Agency Science Tech & Res A multigene assay
US10723794B2 (en) 2015-03-18 2020-07-28 University Of South Carolina Anti-CcL8 antibodies and uses thereof
CN104745697B (en) * 2015-03-24 2018-03-20 济南艾迪康医学检验中心有限公司 Detect the method and primer of NF1 the 31st No. 34 full extron of gene
WO2016196002A1 (en) * 2015-05-29 2016-12-08 The University Of Notre Dame Du Lac Triple negative breast cancer screen and methods of using same in patient treatment selection and risk management
WO2017029215A1 (en) 2015-08-14 2017-02-23 Koninklijke Philips N.V. Assessment of nfkb cellular signaling pathway activity using mathematical modelling of target gene expression
TW201726140A (en) 2015-09-17 2017-08-01 瑞典商阿斯特捷利康公司 Novel biomarkers and methods of treating cancer
EP3363911A4 (en) * 2015-10-14 2019-04-03 Nitto Boseki Co., Ltd Method for determining drug-sensitive human cell lines by analysis method in which measurement of activity of two types of protein kinase is used
US20190080051A1 (en) * 2015-11-11 2019-03-14 Northeastern University Methods And Systems For Profiling Personalized Biomarker Expression Perturbations
KR101847815B1 (en) * 2015-12-17 2018-04-12 바이오코아 주식회사 A method for classification of subtype of triple-negative breast cancer
GB201608000D0 (en) 2016-05-06 2016-06-22 Oxford Biodynamics Ltd Chromosome detection
EP3458610B1 (en) 2016-05-25 2021-05-05 Inbiomotion S.L. Therapeutic treatment of breast cancer based on c-maf status
KR101966493B1 (en) * 2016-06-27 2019-04-05 국립암센터 Biomarkers for Predicting Triple Negative Breast Cancer Prognostication
KR101874716B1 (en) * 2016-12-14 2018-07-04 연세대학교 산학협력단 Methods for classifyng breast cancer subtypes and a device for classifyng breast cancer subtypes using the same
US20200239937A1 (en) * 2017-02-23 2020-07-30 The Council Of The Queensland Institute Of Medical Research Biomarkers for diagnosing conditions
US20210186888A1 (en) * 2017-06-12 2021-06-24 Wake Forest University Health Sciences Nanoparticle compositions for cancer treatment
CA3067585A1 (en) 2017-06-22 2018-12-27 Celgene Corporation Treatment of hepatocellular carcinoma characterized by hepatitis b virus infection
US11654153B2 (en) 2017-11-22 2023-05-23 Inbiomotion S.L. Therapeutic treatment of breast cancer based on c-MAF status
WO2019112966A2 (en) * 2017-12-04 2019-06-13 Nantomics, Llc Subtyping of tnbc and methods
CN108034636B (en) * 2017-12-05 2019-01-11 浙江大学 Human breast cancer cell line and application
KR102211972B1 (en) * 2018-08-02 2021-02-04 엑소젠 피티이. 엘티디 Method for early diagnosis of breast cancer and monitoring after treatment using liquid biopsy multi-cancer gene biomarkers
CN108866197B (en) * 2018-08-23 2021-10-01 中国医学科学院肿瘤医院 Application of PTCH2 gene mutation site in judging susceptibility of young breast cancer
CA3114923A1 (en) * 2018-11-30 2020-06-04 The Trustees Of Indiana University Cancer detection utilizing normal tissue adjacent to breast tumors with genetic ancestry-mapping
CN109913551A (en) * 2019-03-26 2019-06-21 深圳大学 Nucleic acid compositions, breast cancer parting kit and its application method of breast cancer parting
BR102019014302A2 (en) 2019-07-10 2021-12-28 Universidade Federal de Uberlândia TUMOR ANTIBODY BINDING RECOMBINANT PEPTIDES SPECIFIC FOR BREAST CANCER AND USE
CN111948395A (en) * 2020-08-19 2020-11-17 复旦大学附属金山医院 Quadruple marker for diagnosing immune regulation subtype of triple negative breast cancer and application thereof
WO2022051141A1 (en) * 2020-09-03 2022-03-10 The Trustees Of Indiana University Methods for early detection of breast cancer
WO2023204800A1 (en) * 2022-04-19 2023-10-26 Us Oncology Corporate, Inc. Combination therapy
WO2023224487A1 (en) * 2022-05-19 2023-11-23 Agendia N.V. Prediction of response to immune therapy in breast cancer patients
WO2023250397A1 (en) * 2022-06-21 2023-12-28 Bioventures, Llc Methods for the molecular subtyping of tumors from archival tissue
CN115478106B (en) * 2022-08-18 2023-07-07 南方医科大学南方医院 LR-based method for typing triple negative breast cancer and application thereof
WO2024119178A1 (en) * 2022-12-02 2024-06-06 Novelna Inc. Pan-cancer and tissue-specific cancer proteins and methods of use thereof
CN116312802B (en) * 2023-02-01 2023-11-28 中国医学科学院肿瘤医院 Application of characteristic gene TRIM22 in preparation of reagent for regulating and controlling breast cancer related gene expression
CN117607443B (en) * 2024-01-23 2024-04-16 杭州华得森生物技术有限公司 Biomarker combinations for diagnosing breast cancer

Non-Patent Citations (91)

* Cited by examiner, † Cited by third party
Title
Agoff, S.N., et al., "Androgen Receptor Expression in Estrogen Receptor-Negative Breast Cancer", Am J Clin Pathol, 120: 725-731 (2003).
Audeh, M.W., et al., "Phase II Trial of the Oral PARP Inhibitor Olaparib (AZD2281) in BRCA- Deficient Advanced Ovarian Cancer", Journal of Clinnical Oncology, ASCO Annual Meeting Proceedings, 27(15S): 5500 (2009).
Bauer, J.A., et al., "Identification of Markers OfTaxane Sensitivity Using Proteomic and Genomic Analyses of Breast Tumors From Patients Receiving N eoadjuvant Paclitaxel and Radiation", Clin Cancer Res, 16(2): 681-690 (2010).
Bauer, J.A., et al., "RNA Interference (RNAi) Screening Approach Identifies Agents That Enhance Paclitaxel Activity in Breast Cancer Cells", Breast Cancer Research, 12:R41, 16 pages (2010).
Bertucci, F., et al., "Gene Expression Profiling Shows Medullary Breast Cancer is a Subgroup of Basal Breast Cancers", Cancer Res, 66(9): 4636-4644 (2006).
Bertucci, F., et al., "How Baseal Are Triple-Negative Breast Cancers?", Int. J Cancer, 123: 236-240 (2008).
Bhattacharyya, A., et al., "The Breast Cancer-Susceptibility Gene BRCAJ Is Required For Subnuclear Assembly of Rad5 I and Survival Following Treatment With the DNA Crosslinking Agent Cisplatin", J. Biol Chem, 275(31): 23899-23903 (2000).
Birrell, S.N., et al., "Disruption of Androgen Receptor Signaling by Synthetic Progestins May Increase Risk of Developing Breast Cancer", The FASEB Journal, 21: 9 pages (2007).
Boersma, B.J., et al. "A Stromal Gene Signature Associated With Inflammatory Breast Cancer", Int J Cancer, 122: 1324-1332 (2008).
Bos, P.D., et al., "Genes That Mediate Breast Cancer Metastasis to the Brain", Nature, 18:459(7249): 1005-1009 (2009).
Burstein, H.J., et al., "Phase TT Study of Sunitinib Malate, An Oral Multitrageted Tyrosine Kinase Inhibitor, in Patients With Metastatic Breast Cancer Previously Treate With An Anthracycline and a Taxane", J Clin Oneal, 26(11): 1810-1816 (2008).
Carey, L.A., et al., "The Triple Negative Paradox: Primaiy Tumor Chemosensitivity of Breast Cancer Subtypes", Clin Cancer Res, 13(8): 2329-2334 (2007).
Carmeci, C., et al., "Analysis of Estrogen Receptor Messenger RNA in Breast Carcinomas From Archival Specimens is Predictive of Tumor Biology", Am .1 Path, 150(5): 1563-1570 (1997).
Carotenuto, P., et al., "Triple Negative Breast Cancer: From Molecular Portrait to Therapeutic Intervention", Critical Reviews in Eukaryotic Gene Expression, 20: 17-34 (2010).
Carver, B.S., et al., "Reciprocal Feedback Regulation of PI3K and Androgen Receptor Signaling in PTEN-Deficient Prostate Cancer", Cancer Cell, 19: 575-586 (2011).
Chakravarthy, A.B., et al., "Neoadjuvant Concurrent Paclitaxel and Radiation in Stage II/III Breast Cancer", Clin Cancer Res., 12(5): 1570-1576 (2006).
Chen, C.D., et al., "Molecular Determinants of Resistance to Antiandrogen Therapy", Nature Medicine, 10: 33-39 (2004).
Chen, Xi, et al., "TNBCtype: ASubtyping Tool For Triple-Negative Breast Cancer", Cancer Informatics, 11: 147-156 (2012).
Cheung et al. (Nature Genetics 2003 vol. 33 p. 422).
Chin, K., et al., "Genomic and Transcriptional Abberations Linked To Breast Cancer Pathophysiologies", Cancer Cell, 10: 529-541 (2006).
Cobb et al. (Crit Care Med 2002 vol. 30 p. 2711) (Year: 2002). *
D'Andrea, A.D. and Grompe, M., "The Fanconi Anaemia/BRCA Pathway", Nature Reviews Cancer, 3: 23-34 (2003).
Dent, R., et al., "Triple-Negative Breast Cancer: Clinical Features and Patterns of Recurrence", Clin Cancer Res, 13 (15): 4429-4434 (2007).
Desmedt, C., et al., "Strong Time Dependence of the 76-Gene Prognostic Signature For Node- Negative Breast Cancer Patients in the TRANSBIG Multicenter Independent Validation Series", Clin Cancer Res, 13(11 ): 3207-3214 (2007).
Doane, A.S., et al., "An Estrogen Receptor-Negative Breast Cancer Subset Characterized by a Hormonally Regulated Transcriptional Program and Response to Androgen", Oncogene, 25 :3994-4008 (2006).
Ellard, S.L., et al., "Randomized Phase II Study Comparing Two Schedules of Everolimus in Patients With Recurrent/Metastatic Breast Cancer: NCTC Clinical Trials Group TND.163", J Clin Oncol, 27(27): 4536-4541 (2009).
Enard et al. (Science 2002 vol. 296 p. 340).
Evers, B., et al., "Selective Inhibition OfBRCA2-Deficient Mammary Tumor Cell Growth By AZD2281 and Cisplatin", Clin Cancer Res, 14(12): 3916-3925 (2008).
Farmer, H., et al., "Targeting the DNA Repair Defect in BRCA Mutant Cells as a Therapeutic Strategy", Nature, 434: 917-920 (2005).
Farmer, P., et al., "Identification of Molecular Apocrine Breast Tumours by Microarray Analysis", Oncogene, 24: 4660-4671 (2005).
Feschendorff, A.E., et al., "An Immune Response Gene Expression Module Identifies a Good Prognosis Subtype in Estrogen Receptor Negative Breast Cancer", Genome Biology, 8: RI57, 16 pages (2007).
Finn, R.S., et al., "Dasatinib, An Orally Active Small Molecule Inhibitor of Both the src and abl Kinases, Selectively Inhibits Growth of Basal-Type/ "Triple-Negative" Breast Cancer Cell Lines Growing In Vitro", Breast Cancer Res Treat, 105: 319-326 (2007).
Fong, P.C., et al., "Inhibition of Poly(ADP-Ribose) Polymerase in Tumors From BRCA Mutation Carriers", The New England Journal of Medicine, 361(2): 123-134 (2009).
Garber, J.E., et al. "Neo-Adjuvant Cisplatin (CDDP) in "Triple -Negative" Breast Cancer (BC)", Breast Cancer Research and Treatment, 29th Annual San Antonio Breast Cancer Symposium, Dec. 14-17, 100: S149, Abstract#3074 (2006).
Gibson, G.R., et al., "Metaplastic Breast Cancer : Clinical Features and Outcomes", The American Surgeon, 71: 725-730 (2005).
Gonzalez-Angulo, A.M., et al., "Androgen Receptor Levels and Association With PIK3CA Mutations and Prognosis in Breast Cancer", Clin Cancer Res, 15(7): 2472-2478 (2009).
Guarino, M., "Src Signaling in Cancer Invasion", Journal of Cellular Physiology, 223: 14-26 (2010).
Haffty, B.G., et al., "Locoregional Relapse and Distant Metastasis in Conservatively Managed Triple Negative Early-Stage Breast Cancer", J Clin Oneal., 24(36): 5652-5657 (2006).
Hayes, M.J., et al., "Genetic Changes OfWnt Pathyway Genes Are Common Events in Metaplastic Carcinomas of the Breast", Clin Cancer Res, 14(13): 4038-4044 (2008).
Hennessy, B.T., et al., Characterization of a Naturally Occurring Breast Cancer Subset Enriched in Epithelial-To-Mesenchymal Transition and Stem Cell Characteristics, Cancer Res, 69(10): 4116-4124 (2009).
Hoeijmakers, J.H., "Genome Maintenance Mechanisms for Preventing Cancer", Nature, 411: 366-374 (2001).
Hu, Z., et al., "The Molecular Portraits of Breast Tumors Are Conserved Across Microarray Platforms", BMC Genomics, 7:96, 12 pages (2006).
Jones, P., et al., "Discovery Of2-{4-[(3S)-Piperidin-3-yl]phenyl}-2H-indazole-7-carboxamide (MK-4827): A Novel Oral Poly(ADP-ribose)polymerase (PARP) Inhibitor Efficacious in BRCA-1 and -2 Mutant Tumors", J. Med. Chem, 52: 7170-7185 (2009).
Juul, N., et al., "Assessment of an RNA Interference Screen-Derived Mitotic and Ceramide Pathyway Metagene as a Predictor of Response to Neoadjuvant Paclitaxel for Primmy Tripl-Negative Breast Cancer: A Retrospective Analysis of Five Clinical Trials", The Lancet, 11: 358-365 (2010).
Kam, T., et al., "Data Driven Derivation of Cutoffs From a Pool of 3,030 Affymetrix Anays to STratify Distinct Clinical Types of Breast Cancer", Breast Cancer Res Treat, 120: 567-579 (2010).
Kenny, P.A., et al., "The Morphologies of Breast Cancer Cell Lines in Three-Dimensional Assays Conelate With Their Profiles of Gene Expression", Molecular Oncology, I: 84-96 (2007).
Kreike, B., et al., "Gene Expression Profiling and Histopathological Characterzation of Triple- Negative/Basal-Like Breast Carcinomas", Breast Cancer Research, 9: R65, 14 pages (2007).
Kwei, K.A., et al., "Genomic Instability in Breast Cancer: Pathogenesis and Clinical Implications", Mal. Oneal, 4(3): 255-266 (2010).
Lehmann, B.D., et al., "Identification of Human Triple-Negative Breast Cancer Subtypes and Preclinical Models for Selection of Targeted Therapies", Journal of Clinical Investigation, 121(7): 2750-2767 (2011).
Li, Y., et al., "Amplification OfLAPTM4B and YWHAZ Contributes to Chemotherapy Resistance and Recunence of Breast Cancer", Nature Medicine, 16(2): 214-218 (2010).
Liu, X., et al., "Simultaneous Targeting of the Androgen Receptor and PI3K/mTOR Pathway in Androgen-Dependent and Androgen-Independent Prostate Cancer Cells", Journal of Clinical Oncology, ASCO Annual Meeting Abstracts, 28 (15_suppl):c15049, 1 page (2010).
MacDonald, B.T., et al., "Wnt/B-Catenin Signaling: Components, Mechanisms, and Disease", Dev. Cell, 17: 9-26 (2009).
Maiiy, B., et al., "Frequent PTEN Genomic Alterations and Activated Phosphatidylinositol 3- Kinase Pathyway in Basal-Like Breast Cancer Cells", Breast Cancer Research, 10: RIOI, 15 pages (2008).
Maira, S.M., et al., "Identification and Characterization OF NVP-BEZ235, A New Orally Available Dual Phosphatidylinositol 3-Kinase/Mammalian Target of Rapamycin Inhibitor With Potentln Vivo Antitumor Activity", Mal Can Ther, 7(7): 1851-1863 (2008).
Mani, S.A., et al., "The Epithclial-Mcscnchymal Transition Generates Cells With Properties of Stem Cells", Cell, 133:705-715 (2008).
Mayer, I.A., et al., "A Phase II Neoadjuvant Study of Cisplatin/Paclitaxel With or Without RADOOI in Patients With Triple-Negative (TN) Locally Advanced Breast Cancer (BC)", J. Clin Oneal, 28: 15s (Suppl; abstract TPSI 19) (2010).
Miller, L.D., et al., "An Expression Signature For p53 Status in Human Breast Cancer Predicts Mutation Status, Transcriptional Effects, and Patient Survival", PNAS, 102(38): 13550-13555 (2005).
Minn, A.J., et al., "Genes That Mediate Breast Cancer Metastasis to Lung", Nature, 436: 518-524 (2005).
Morris, G.J., et al., "Differences in Breast Carcinoma Characteristics in Newly Diagnosed African-American and Caucasian Patients", Cancer, 110: 876-884 (2007).
Neve, R.M., et al., "A Collection of Breast Cancer Cell Lines for the Study of Functionally Distinct Cancer Subtypes", Cancer Cell, 10: 515-527 (2006).
Newton et al. (Journal of Computational Biology 2001 vol. 8 p. 37).
Nielsen, T.O., et al., "Immunohistochemical and Clinical Characterization of the Basal-Like Subtype OfInvasive Breast Carcinoma", Clinical Cancer Research, 10: 5367-5374 (2004).
Notification Concerning Trnnsmittal of International Preliminary Report on Patentability for PCT/US2012/065724, Markers of Triple-Negative Breast Cancer and Uses Thereof, dated May 30, 2004. (4783.1000-001).
Notification of Transmittal of the International Search Report and the Written Opinion of the International Searching Authority for PCT/US2012/065724, Markers of Triple-Negative Breast Cancer and Uses Thereof, dated Feb. 7, 2013. (4783.1000-001).
Pal, S.K., et al., "Triple Negative Breast Cancer: Unmet Medical Needs", Breast Cancer Res Treat, 125:627-636 (2011).
Pawitan, Y., et al., "Gene Expression Profiling Spares Early Breast Cancer Patients From Adjuvant Therapy: Derived and Validated in Two Population-Based Cohorts", Breast Cancer Research, 7: R953-R964 (2005).
Pegram., M.D., et al., "Phase II Study of Receptor-Enhanced Chemosensitivity Using Recombinant Humanized Anti-p185HER/neu Monoclonal Antibody Plus Cisplatin 1n Patients With HER2/neu-Overexpressing Metastatic Breast Cancer Refractory to Chemotherapy Treatment", Journal of Clinical Oncology, 16(8): 2659-2671 (1998).
Prat, A., et al., "Phenotypic and Molecular Characterization of the Claudin-Low Intrinsic Subtype of Breast Cancer", Breast Cancer Research, 12: R68, 18 pages, (2010).
Press, M.F., et al., "HER-2 GcncAmplification, HER-2 and Epidermal Growth Factor Receptor mRNA and Protein Expression, and Lapatinib Efficacy in Women With Metastatic Breast Cancer", Clincal Cancer Research, 14(23): 7861-7870 (2008).
Rakha, E. and Ellis, I., "Are Triple-Negative and Basal-Like Breast Cancer Synonymous?", Clinical Cancer Research, 14(2): 618 (2008).
Rakha, E.A., et al., "Triple-Negative Breast Cancer: Distinguishing Between Basal and Nonbasal Subtypes", Clinical Cancer Research, 15(7): 2302-2310 (2009).
Richardson, A.L., et al., "X Chromosomal Abnormalities in Basal-Like Human Breast Cancer", Cancer Cell, 9: 121-132 (2006).
Samuels, Y., et al., "High Frequency of Mutations of the PIK3CA Gene in Human Cancers", Science, 304: 554 (2004).
Schmidt, M., et al., "The Humoral Immune System Has a Key Prognostic Impact in Node- Negative Breast Cancer", Cancer Research, 68(13): 5405-5413 (2008).
Shin, S.Y., et al., "Functional Roles of Multiple Feedback Loops in Extracellular Signal-Regulated Kinase and Wnt Signaling Pathways That Regulate Epithelial-Mesenchymal Transition", Cancer Research, 70(17): 6715-6724 (2010).
Shipitsin, M., et al., "Molecular Definition of Breast Tumor Heterogeneity", Cancer Cell, 11: 259-273 (2007).
Silver, D.P., et al., "Efficacy of Neoadjuvant Cisplatin in Triple-Negative Breast Cancer", Journal OfClinical Oncology, 28(7): 1145-1153 (2010).
Solit, D.B., et al., "I 7-Allylamino-17-Demethoxygeldanamycin Induces the Degradation of Androgen Receptor and HER-2/neu and Inhibits the Growth of Prostate Cancer Xenografts", Clinical Cancer Research, 8: 986-993 (2002).
Sotiriou, C., et al., "Gene Expression Profiling in Breast Cancer: Understanding the Molecular Basis OfHistologic Grade to Improve Prognosis", Journal of The National Cancer Institute, 98(4): 262-272 (2006).
Stefansson, O.A., et al., "Genomic Profiling of Breast Tumours 1n Relation To BRCA Abnormalities and Phenotypes", Breast Cancer Research, 11: R4 7, 14 pages (2009).
Stemke-Hale, K., et al., "An Integrative Genomic and Proteomic Analysis of PIK3CA, PTEN, and AKT Mutations in Breast Cancer", Cancer Research, 68(15): 6084-6091 (2008).
Subramanian, A., et al., "Gene Set Enrichment Analysis: A Knowledge-BAsed Approach for Interpreting Genome-Wide Expression Profiles", PNAS, 102(43): 15545-15550 (2005).
Telli, M.L and Ford, J.M., "Novel Treatment Approaches for Triple-Negative Breast Cancer", Clinical Breast Cancer, IO (Supp 1): EI6-E22 (2010).
Toyama et al. (BMC Cancer Oct. 25, 2008 vol. 8 p. 309).
Uhm et al. (Int J Cancer 2009 vol. 124 p. 1457).
Wang, Y., et al., "Gene-Expression Profiles to Predict Distant Metastasis of Lymph-Node-Negative Primary Breast Cancer", The Lancet, 365: 671-679 (2005).
Wiggans, R.G., et al., "Phase-II Trial OfTamoxifen in Advanced Breast Cancer", Cancer Chemother Pharmacol, 3: 45-48 (1979).
Wu (Journal of pathology 2001 vol. 195 p. 53).
Yang, X.R., et al., "Associations of Breast Cancer Risk Factors With Tumor Subtypes: A Pooled Anaylsis From the Breast Cancer Association Consortium Studies", J National Cancer Institute, 103(3): 250-263 (2011).
Yu, K, et al., "A Precisely Regulated Gene Expression Cassette Potently Modulates Metastasis and Survival in Multiple Solid Cancers", PLOS Genetics, 4(7): eI 000129, 12 pages (2008).
Zhou, X. and Agazie, Y., "Abstract #3388 SHP2 Promotes Breast Cancer Development", Proc. Am Asso Cancer Res; Apr. 18-22, 2009; Denver, CO: AACR; 2009.

Also Published As

Publication number Publication date
US20200224282A1 (en) 2020-07-16
AU2012340186A1 (en) 2014-06-19
CA2856295A1 (en) 2013-05-23
WO2013075059A1 (en) 2013-05-23
EP2780469A1 (en) 2014-09-24
US20140303133A1 (en) 2014-10-09
US20240158865A1 (en) 2024-05-16

Similar Documents

Publication Publication Date Title
US11788147B2 (en) Markers of triple-negative breast cancer and uses thereof
Lehmann et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies
Kharma et al. STAT1 drives tumor progression in serous papillary endometrial cancer
Sawayama et al. Investigation of colorectal cancer in accordance with consensus molecular subtype classification
Wong et al. Poor survival with wild-type TP53 ovarian cancer?
WO2013052480A1 (en) Marker-based prognostic risk score in colon cancer
US11015190B2 (en) Method of treating a patient having renal cancer
JP2017506506A (en) Molecular diagnostic tests for response to anti-angiogenic drugs and prediction of cancer prognosis
US20140155397A1 (en) Emt signatures and predictive markers and method of using the same
WO2013134649A1 (en) Biomarkers for cancer stem cells and related methods of use
US11851712B2 (en) Replication stress response biomarkers for immunotherapy response
Chen et al. Peripheral immune cell gene expression changes in advanced non-small cell lung cancer patients treated with first line combination chemotherapy
Cejalvo et al. Clinical implications of routine genomic mutation sequencing in PIK3CA/AKT1 and KRAS/NRAS/BRAF in metastatic breast cancer
Minna et al. Tumor mRNA expression profiles predict responses to chemotherapy
Su et al. DNA methylome and transcriptome landscapes of cancer-associated fibroblasts reveal a smoking-associated malignancy index
Sattar et al. Diagnostic and prognostic biomarkers in colorectal cancer and the potential role of exosomes in drug delivery
Rijnders et al. Anti–PD-1 Efficacy in Patients with Metastatic Urothelial Cancer Associates with Intratumoral Juxtaposition of T Helper-Type 1 and CD8+ T cells
Chang et al. Digital spatial profiling to predict recurrence in grade 3 stage I lung adenocarcinoma
Ohara et al. The evolution of genomic, transcriptomic, and single-cell protein markers of metastatic upper tract urothelial carcinoma
Cejalvo Andújar Uncovering the molecular and cellular mechanisms of metastatic dormancy in luminal breast cancer
Tsai et al. A serum-induced transcriptome and serum cytokine signature obtained at diagnosis correlates with the development of early pancreatic ductal adenocarcinoma metastasis
US20230257825A1 (en) Breast cancer biomarkers and methods of use
Chamdin et al. Bcl6 is expressed in neuroblastoma: tumor cell type-specific expression predicts outcome
EP3665307B1 (en) Materials and methods for stratifying and treating cancers
이경열 Clinicopathologic significance of CEP17 copy number gain in breast cancer

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

AS Assignment

Owner name: VANDERBILT UNIVERSITY, TENNESSEE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PIETENPOL, JENNIFER A;LEHMANN, BRIAN;BAUER, JOSH;AND OTHERS;SIGNING DATES FROM 20141021 TO 20141027;REEL/FRAME:052409/0072

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

STCC Information on status: application revival

Free format text: WITHDRAWN ABANDONMENT, AWAITING EXAMINER ACTION

STPP Information on status: patent application and granting procedure in general

Free format text: AMENDMENT AFTER NOTICE OF APPEAL

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STPP Information on status: patent application and granting procedure in general

Free format text: AWAITING TC RESP, ISSUE FEE PAYMENT VERIFIED

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE